You are on page 1of 26

Interdiscip Toxicol. 2019; Vol. 12(2): 45–70.

interdisciplinary
doi: 10.2478/intox-2019-0007

Copyright © 2019 SETOX & IEPT CEM SASc.


This is an Open Access article distributed under the terms of the Creative Commons Attri-
bution-NonCommercial-NoDerivatives 4.0 License (https://creativecommons.org/licenses/
by-nc-nd/4.0).

REVIEW ARTICLE

Aluminium toxicosis: a review of


toxic actions and effects
Ikechukwu Onyebuchi IGBOKWE 1, Ephraim IGWENAGU 1, Nanacha Afifi IGBOKWE 2
1 Department of Veterinary Pathology, Faculty of Veterinary Medicine, University of Maiduguri, Maiduguri, Nigeria
2 Department Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Maiduguri, Maiduguri, Nigeria

ITX120219A01  •  Received: 22 November 2018  •  Accepted: 29 August 2019

ABSTRACT
Aluminium (Al) is frequently accessible to animal and human populations to the extent that intoxications may occur. Intake of Al is
by inhalation of aerosols or particles, ingestion of food, water and medicaments, skin contact, vaccination, dialysis and infusions.
Toxic actions of Al induce oxidative stress, immunologic alterations, genotoxicity, pro-inflammatory effect, peptide denaturation or
transformation, enzymatic dysfunction, metabolic derangement, amyloidogenesis, membrane perturbation, iron dyshomeostasis,
apoptosis, necrosis and dysplasia. The pathological conditions associated with Al toxicosis are desquamative interstitial pneumonia,
pulmonary alveolar proteinosis, granulomas, granulomatosis and fibrosis, toxic myocarditis, thrombosis and ischemic stroke, granulo-
matous enteritis, Crohn’s disease, inflammatory bowel diseases, anemia, Alzheimer’s disease, dementia, sclerosis, autism, macrophagic
myofasciitis, osteomalacia, oligospermia and infertility, hepatorenal disease, breast cancer and cyst, pancreatitis, pancreatic necrosis
and diabetes mellitus. The review provides a broad overview of Al toxicosis as a background for sustained investigations of the
toxicology of Al compounds of public health importance.

KEY WORDS: aluminium; intoxication; pathology; toxicity; toxicosis

Introduction

Aluminium (Al) is the most widely distributed metal in to high levels of Al (Sorgdrager et al., 1998; Vandenplas et
the environment (Delhaize and Ryan, 1995; Ranjbar et al., al., 1998; Boran et al., 2013).
2008; Exley and House, 2011) occurring naturally in the Several chemical compounds with Al are in extensive
trivalent state (Al+3) as silicates, oxides and hydroxides, use in various products and processes associated with
but may combine with other elements such as chlorine, human activities. These compounds are Al chloride, Al
sulphur, fluorine, as well as form complexes with organic hydroxide (alumina trihyrate), Al nitrate, Al phosphate,
matter (Jones and Bennet, 1986; Ganrot, 1986; Martin, Al sulfate (alum), Al potassium (potash alum), Al ammo-
1992). Environmental media may be contaminated by Al nium sulfate (ammonium alum) and Al silicate (Anon,
from anthropogenic sources and through the weathering 1982; Lewis, 2001). The compounds are used in crude
of rocks and minerals. Weathering processes on rocks oil refining and cracking of petroleum; manufacturing
release more Al to the environment than human-related of cooking utensils and foils, parchment paper, printing
activities (Lantzy and MacKenzie, 1979). Exposures to ink, glass, ceramics, pottery, incandescent filaments,
Al occur in occupations associated with mining and fireworks, explosives, photographic flashlight, electric
processing of ore, scrap metal recycling, deployment insulators, cement, paints and varnishes, fumigants and
and use of Al-containing compounds and products, and pesticides, lubricants, detergents, cosmetics, pharma-
during engagement in Al metal cutting, sawing, filing and ceuticals (drugs), vaccines, as well as in water treatment
welding. Animals and humans living in environments and purification, treating sewage and fur, tanning leather,
contaminated by industrial wastes may also be exposed waterproofing clothes and concretes, industrial filtration,
hemodialysis, measuring radiation exposure, in products
as flame retardant and fireproofing, anticorrosion agent,
food additives to prevent caking as well as components
Correspondence address: of baking powders and colorants (Anon, 1982, 2008a;
Ikechukwu Onyebuchi Igbokwe, DVM, MVSc, PhD, FCVSN Malakoff, 2000; Lewis, 2001; Soni et al., 2001; Saiyed and
Faculty of Veterinary Medicine, University of Maiduguri, Yokel, 2005).
Strategic Animal Research Group
P O Box 8000, Maiduguri, Nigeria The Al ion has no physiological role in metabolic
e-mail: ikeigbokwe@gmail.com; ikeigbokwe@unimaid.edu.ng processes (Exley and House, 2011) but it can be a metallic
46 Aluminium toxicosis
Ikechukwu Onyebuchi Igbokwe, Ephraim Igwenagu, Nanacha Afifi Igbokwe

toxicant to humans and animals (Becaria et al., 2002) discovered to have focused on Al toxicity of one system
when there is high body burden of the metal after natural of the body in each review, covering the scope of nervous,
or unnatural exposure (Exley, 2013). Al was considered reproductive, respiratory, mammary, skeletal and immune
unsafe to humans after the discovery of increased levels tissue toxicities. One review addressed the toxicities in
of Al in brain tissues of patients with encephalopathy, bone, hematopoietic tissue and kidney (Jeffrey et al., 1996)
having been exposed to Al accumulation through dialysis and another summarized the physiological alterations in
(Alfrey and Solomons, 1976). Toxicosis due to Al accumu- the musculoskeletal, respiratory, cardiovascular, hepatobi-
lation in mammalian tissues was associated with various liary, endocrine, urinary and reproductive systems(Nayak,
pathologic effects (Wills and Savory, 1983; Kaiser et al., 2000). Thus the question raised was whether Al toxicosis,
1984; Boyce et al., 1986; Drüeke et al., 1986; Hewitt et al., as a disease entity, existed in the literature with cur-
1990; Bushinsky et al., 1995; Reinke et al., 2003; Abubakar rent research information. The literature search for Al
et al., 2004; Bogdanović et al., 2008; Yousef and Salama, toxicosis as a narrative review (Green et al., 2006) with
2009; Khattab et al., 2010; Blaylock, 2012; Buraimoh and a broad thematic approach was unproductive and this
Ojo, 2013; Sumathi et al., 2013). Recent reviews on toxic observation justified the need for the current review.
effects of Al covered reproductive toxicity (Mouro et al., Toxicosis associated with Al exposure is the pathologi-
2017), pulmonary lesions (Kongerud and Søyseth, 2014; cal condition or disease caused by the toxic actions of Al
Taiwo, 2014), impact on the breast (Darbre, 2016), bone and its compounds. The literature search was intended
abnormalities (Chappard et al., 2016; Klein, 2019), immu- to collate, synthesize and integrate the published reports
notoxity (Zhu et al., 2014a) and neurologic disorders on the subject matter without meta-analysis and critical
(Colomina and Peris-Sampedro, 2017; Morris et al., 2017). evaluation of published data. For this review, the major
This review is an abridged and global overview of toxic themes for the literature search under the title included
effects of Al and its compounds, covering some relevant exposure modalities, toxic actions and effects in cells,
aspects of exposure and updated systemic toxicosis in tissues and systems of the body (Figure 1). These themes
humans and animals, relevant as background for prospec- provided the key words and phrases for the internet search.
tive toxicopathologic studies. The initial search platform was usually GOOGLE.COM
with the linked GOOGLE SCHOLAR helping to search
related articles. Subsequently, the search was extended to
Literature search justification and methods MEDLINE, PUBMED, PMC Europe, RESEARCHGATE,
SCOPUS, SCIENCEDIRECT, SAGE, TANDFONLINE
The initial goal in our study group was to explore the and SPRINGERLINK. On each platform, an article that
role of the ubiquitous Al ion in erythrocyte membrane was found could also have links to related articles and
dysfunction (Igbokwe, 2016) and metabolic dysregulation these links were followed to enrich the search outcomes.
(Igwenagu, 2017). With the preliminary literature search Every article was read as an abstract or full article after
starting in 2013 and looking backwards in time, research downloading to the literature bank. At this stage, “back-
publications revealed a myriad of toxic actions of Al caus- ward search” based on references of read articles could
ing pathological conditions. Several narrative literature be done and “forward search” was required when new
reviews, referred to in the introductory section, were themes emerged that needed further exploration. The

Figure 1. Major themes for the literature search on aluminium toxicosis.

ISSN: 1337-6853 (print version) | 1337-9569 (electronic version)


Interdisciplinary Toxicology. 2019; Vol. 12(2): 45–70 47
Full-text also available online on PubMed Central

integration of the literature search processes are illus-


trated in Figure 2. Occasionally, article request options Literature
were used to obtain restricted publications and the articles Search: HOW
were received from authors. The literature collated from
the search was read for content comprehension. The body
Research Articles:
of knowledge was summarized and narrated after cogni- Backward
Primary (original research)
Forward
Search Search
tive reflection and integration to represent the current Secondary (review)

knowledge of the literature on Al toxicosis. The articles


with thematic contents were included for the review when
GOOGLE.COM
they were published in journals with reputable standing.
The contents of the excluded articles were peripheral to
the themes under review. MEDLINE
PUBMED
PMC Europe
RESEARCHGATE
Exposure to aluminium
SCOPUS
SCIENCEDIRECT
Aluminium intake
SAGE
Aluminium in the air TANDFONLINE
The largest source of airborne Al-containing particles SPRINGERLINK
is the dust from soil and rocks (Lee and Von Lehmden,
1973; Sorenson et al., 1974). Human activities, such as
Figure 2. Integrated literature search process.
mining and agriculture, contribute to the dust in winds
(Eisenreich, 1980; Filipek et al., 1987). About 13% of
atmospheric Al is attributed to anthropogenic emissions
(Lantzy and MacKenzie, 1979). The major anthropogenic
sources of Al-containing particulate matter include coal 1985; Henshaw et al., 1993; Cech and Montera, 2000). In
combustion, Al production, iron and steel foundries, pure water, Al has a minimum solubility in the pH range
brass and bronze refineries, motor vehicle emissions and of 5.5–6.0 and concentrations of dissolved Al increase
other industrial activities such as smelting, filing, sawing, at higher or lower pH values (Browne et al., 1990). The
welding of Al metals (Lee and Von Lehmden, 1973; Ondov source of water for human and animal consumption and
et al., 1982; Que Hee et al., 1982). Cigarette smoke may the purification process involved may influence the Al
contribute to the concentration of Al in the air (Exley content of drinking water as source of exposure.
et al., 2006; Kazi et al., 2009; Pappas, 2011; Afridi et al.,
2015). The air containing Al particles or droplets becomes Aluminium in food
the source of Al in inhaled aerosols. Al is present in foods naturally or from the use of
Al-containing food additives (Sepe et al., 2001; Flaten,
Aluminium in drinking water 2002; Hayacibara et al., 2004; Yokel et al., 2008). The
Al occurs ubiquitously in natural waters due to weather- concentrations in foods and beverages vary widely,
ing of Al-containing rocks and minerals and mobilization depending upon the food product, the type of processing
from terrestrial to aquatic environment (Campbell et al., used, and the geographical areas in which the food crops
1992). This mobilization of Al is often seasonal in nature are grown (Sorenson et al., 1974; Pennington and Schoen,
and is associated with pH depressions (acidification) 1995). The foods highest in Al are those that contain Al
occurring during the spring snow melt or associated with additives (Pennington, 1988; Greger, 1992; Saiyed and
erosion from specific storm events (Rosseland et al., 1990; Yokel, 2005; Yokel and Florence, 2006; Yokel, 2012). The
Nelson and Campbell, 1991; Campbell et al., 1992). Al use of Al cookware, utensils and wrappings can increase
concentrations in surface waters can be increased directly the amount of Al in food (Liukkonen-Lilja and Piepponen
or indirectly by human activities through industrial and 1992; Pennington and Schoen, 1995). The migration of Al
municipal discharges, surface run-off, tributary inflow, from cookware into food increases with the acidity of the
groundwater seepage, and wet and dry atmospheric depo- food and the duration of exposure (Valkonen and Aitio,
sition (Eisenreich, 1980). Industrial release of Al in waste 1997; Lin et al. 1997). Al was also reported to migrate into
materials into surface waters from processing and manu- fish grilled on Al foil and the migration of Al into foods
facturing facilities could be toxic to aquatic life (Filipek appeared to be dependent on factors such as temperature,
et al., 1987; Trieff et al., 1995; His et al., 1996; Gensemer duration of cooking, the composition and pH of the food,
and Playle, 1999). Acidic drainage from mines or acid and the presence of other substances like organic acids
rain may cause an increase in the dissolved Al content and salts (Ranau et al., 2001). Foods found to be naturally
of the surrounding water bodies (Cronan and Schofield, high in Al include potatoes, spinach and tea (Pennington
1979; Filipek et al., 1987). The use of Al compounds as and Schoen, 1995; Stahl et al., 2011). Processed dairy
coagulating agents in the treatment of water for drinking products and flour may be high in Al if they contain
could increase its Al content (Qureshi and Malmberg, Al-based food additives (Pennington and Schoen, 1995).

Copyright © 2019 SETOX & Institute of Experimental Pharmacology and Toxicology, CEM SASc.
48 Aluminium toxicosis
Ikechukwu Onyebuchi Igbokwe, Ephraim Igwenagu, Nanacha Afifi Igbokwe

Daily intakes of Al in humans from food range from 3.4 of Al per tablet, capsule or 5 ml of suspension (Zhou
to 9 mg/day (Pennington and Schoen, 1995; Biego et al., and Yokel, 2005). The use of other consumer items
1998; Yang et al., 2014). It is unlikely that Al-containing such as dentifrices, disinfectants, fumigants, pesticides,
food additives are intentionally added to the diets of live- anti-perspirants and some cosmetics are sources of Al
stock and pets yet, Al contamination of some additives exposure (Lewis, 2001; Pineau et al., 2014). Al hydroxide,
used in livestock and pet food is possible (Burgoin, 1992). Al phosphate, Al potassium sulfate (alum), and Al silicate
Thus Al contents of harvested food products, processed (zeolite) are used in the preparation of a number of vac-
foods, and cooked, baked or grilled foods may be sources cines to adsorb antigenic components and to serve as
of Al exposure. adjuvant that enhance immune response (Lione, 1985;
Tomljenovic and Shaw, 2011; Issa et al., 2014). Adjuvant
Aluminium in pharmaceuticals and agrochemicals as a source of Al during vaccinations has been receiving
The route of intoxications with pharmaceuticals and agro- attention in research (Malakoff, 2000; Keith et al., 2002;
chemical sources may be through inhalation of aerosols, Mitkus et al., 2011; Glanz et al., 2015) and it is presumed
ingestion of medications or by parenteral administration. that there could be mistakes in adjusting Al content of
Humans and animals are exposed to Al-containing vaccines to body weights of neonates who stand the risk
medications such as phosphate binders, antacids, buffered of Al toxicity from vaccines (Lyons-Weiler and Ricketson,
analgesics, antidiarrheal and antiulcer drugs (Lione, 1983, 2018). More Al was absorbed into blood by rabbits after
1985; Yokel and McNamara, 2001; Krewski et al., 2007). intramuscular injection with adjuvant containing Al
Various intravenously administered pharmaceutical phosphate compared to Al hydroxide (Hem, 2002). It is
products were reported to contain 684–5977 µg/g of Al unlikely that parenteral Al administrations are a major
(Sedman et al., 1985). Many antacids contain 104–208 mg source of Al exposure to livestock or pets (Issa et al., 2014).

Aluminium Compounds

Aerosol Infusions
Vaccines
Water Dialysate
Food
INHALATION Medicaments

LUNG
INGESTION PARENTERAL TREATMENT
?

Decreased by Increasde by
ABSORPTION
• Iron overload • Chronic kidney disease
• Dietary • Deficiency of Ca & Mg ions
- Phytates • Dietary
- Polyphenols - Citrate
- Phosphate
BLOOD CIRCULATION
- Maltol
- Silicon - Lactate
- Sialic acid - Fluoride

Urine
Bile
Faeces EXCRETION TISSUE ACCUMULATION
Sweat
Hairs
Nail Increased by chelators
Sebum
Semen
• Deferoxamine
Milk • Malic acid
• Citric acid
• Malonic acid
• Oxalic acid
• Succinic acid

Figure 3. Factors affecting tissue accumulation of aluminium and development of toxicosis.

ISSN: 1337-6853 (print version) | 1337-9569 (electronic version)


Interdisciplinary Toxicology. 2019; Vol. 12(2): 45–70 49
Full-text also available online on PubMed Central

Food or water for livestock could be contaminated with Al About 90% of the Al circulating in the blood is trans-
when Al sulfate and zeolite are applied to litter and waste ported bound to transferrin (iron-transporter protein),
lagoons to reduce phosphorus loss from lands fertilized while the rest of Al binds to albumin and citrate in
with the wastes and to reduce ammonia fumes in facili- the blood (Day et al., 1991; Harris and Messori, 2002;
ties (Moore et al., 1999; Moore et al., 2000; Codling et al., Hemadi et al., 2003; Chen et al., 2010). Cellular uptake
2002). Alum has also been added to dairy slurry to reduce of Al in tissues is relatively slow and is presumed to
ammonia emissions (Lefcourt and Mesinger, 2001). Thus, be mediated by endocytosis and intracellular transfer
this section indicates that Al exposure can arise when of the Al bound to transferrin (Hemadi et al., 2003).
certain pharmaceutical products are administered orally However, Al-transferrin complex may not bind to the
or parenterally to individuals or when agrochemicals transferrin-receptor (Hemadi et al., 2003; Sakajiri et al.,
contaminate food/feed and water taken by individuals or 2010), indicating the existence of an alternative mecha-
those in close proximity inhale aerosols from agrochemi- nism of cellular uptake of Al (DeVoto and Yokel, 1994;
cal fumigants and sprays. Anon, 2011). The total body burden of Al in healthy
humans has been reported to be approximately 30–50
Absorption, distribution and elimination of aluminium mg/kg body weight and normal levels of Al in serum are
The dynamic chain of Al intake, absorption and elimi- approximately 1–3μg/L (Krewski et al., 2007). The mean
nation determines the level of tissue accumulation and serum Al level in 44 non-exposed persons who did not
development of toxicosis (Figure 3). Inhalation and inges- use antacids was reported to be 1.6 μg/L (Valkonen and
tion (via food and water) are the two main routes through Aitio, 1997) and Chen et al. (2010) reported that values
which Al gets into the body (Alfrey, 1980; Teraoka, 1981; in hemodialysis patients were ten-fold higher than the
Jouhanneau et al., 1997). Following inhalation, Al com- values in unexposed individuals. About one-half of the
pounds are deposited in the lungs (Christie et al., 1963; total body Al is in the skeleton, and the levels in human
Stone et al., 1979; Thomson et al., 1986). The lungs con- bone tissue range from 5 to 10 mg/kg (Anon, 2008c). Al
tinually receive Al mostly as particles of Al silicates and has also been found in human skin, lower gastrointes-
other poorly soluble compounds (Thomson et al., 1986). tinal tract, lymph nodes, adrenals, parathyroid glands,
The concentration of Al in the lungs tends to increase and in most soft tissue organs (Anon, 2008b). In rats,
with age and may result in respiratory anomalies where accumulation of Al after oral exposure was higher in the
the Al is localized (Alfrey, 1980; Teraoka, 1981; Taiwo, spleen, liver, bone, and kidneys than in the brain, muscle,
2014). There is no available evidence in literature that heart, or lungs (Anon, 2008b). It has also been reported
particulate or soluble Al gets into the blood circulation that Al can reach the placenta and fetus and to some
from the lungs to be subsequently distributed to other extent distribute to the milk of lactating mothers (Anon,
organs of the body. 2008b). Al levels increase with age in tissues and organs
Gastrointestinal absorption, after ingestion, is the (bone, muscle, lung, liver, and kidney) of experimental
main route through which Al is systemically accumulated animals (Krewski et al., 2007). Moreover, Al has been
in animals and humans, and absorption occurs largely in shown to rapidly enter the brain, extracellular fluid and
the duodenum (Feinroth et al., 1984; Steinhausen et al., the cerebrospinal fluid, with smaller concentrations in
2004). The absorption of Al is usually low and varied when these organs than in the blood (Martin, 1992; Krewski et
compared with the amount ingested (Kawahara et al., al., 2007). The iron status is negatively correlated with
2007). The uptake of Al through gastrointestinal pathway Al accumulation in tissues and animal experiments
is complex and is influenced by various factors including have shown that calcium and magnesium deficiency may
individual differences, age, pH, stomach contents and contribute to accumulation of Al in the brain and bone
type of Al compound (Priest et al., 1996). Al absorption (Anon, 2011).
from water intake (about 0.3%) is greater than from food The Al ion in blood circulation is eliminated primarily
(about 0.1%) (Martyn et al., 1989; Steinhausen et al., 2004; by the kidneys (about 95%) in the urine, presumably as
Anon, 2008b; Zhou et al., 2008).This was attributed to Al citrate (Shirley and Lote, 2005; Krewski et al., 2007;
organic ligands in foods such as phytates and polyphenols Anon, 2008c). Tissue accumulation of Al is reduced by
that were suggested to form complexes with Al ion and citrates and fluorides through renal excretion when the
inhibit its absorption (Reto et al., 2007). Absorption of transferrin-Al binding capacity of the blood is exceeded
Al via the gastrointestinal tract can be enhanced in the (Anon, 2008b). Al is also excreted in the milk, bile, feces,
presence of citrate, maltol, lactate and fluoride in water sweat, hairs, nails, sebum and semen (Gorsky et al.,
or food, and during chronic renal diseases, while the 1979; Greger and Sutherland, 1997; Exley, 2013). Urinary
absorption is reduced in individuals with iron overload, excretion of Al is enhanced by chemical chelators such as
or when ingested with phosphate, silicon, polyphenols deferoxamine and malic, malonic, citric, oxalic and suc-
and sialic acid (Brown et al., 1987; Edwardson et al., cinic acids, as reviewed in the later section on treatment
1993; Anon, 2008c; Zhou et al., 2008). However, there is of Al in this document. On the whole, it is noted that Al
complete Al uptake from parenteral fluids and vaccines accumulation, which is responsible for Al toxicosis, is
with subsequent distribution to various parts of the body enhanced by exposure to Al and its continuous intake,
(Tomljenovic and Shaw, 2011). as well as increased intestinal absorption and decreased
excretion of the metal (Figure 3).

Copyright © 2019 SETOX & Institute of Experimental Pharmacology and Toxicology, CEM SASc.
50 Aluminium toxicosis
Ikechukwu Onyebuchi Igbokwe, Ephraim Igwenagu, Nanacha Afifi Igbokwe

Toxic actions of aluminium The aggregation and precipitation of β-amyloid is trig-


gered and potentiated by Al exposure which is associated
The toxic actions of Al responsible for the toxic effects with Alzheimer’s disease (Bondy and Truong, 1999; Zatta
of the toxicities are diverse and capable of causing a et al., 2005; Exley, 2006) and this phenomenon may be
multifaceted systemic toxicosis. These toxic actions are responsible for neuritic plaque deposition, neuronal
summarized in Table 1. The molecular targets of action death and dysneurogenesis. Fibrillation and aggregation
generate outcomes in the cell and disrupt cellular homeo- of human islet amyloid polypeptide hormone (amylin)
stasis with consequences that lead to lesions in the cell was stimulated by Al exposure leading to formation of
(Figure 4), which are responsible for systemic toxicosis β-pleated sheet structure (Mirhashemi and Aarabi, 2011;
associated with structural and functional abnormalities Mirhashemi and Shahabaddin, 2011; Xu et al., 2016),
of organs. which may predispose pancreatic β-cell to damage. The
Toxic effects of Al arise mainly from its pro-oxidant proteolytic degradation of the amyloid peptides is also
activity which results in oxidative stress, free radical prevented by Al, thereby enhancing the accumulation of
attack and oxidation of cellular proteins and lipids (Exley, the amyloid (Sakamoto et al., 2006). Extracellular sur-
2013). Protein polypeptides are transformed to second- faces and intracellular ligands may likely associate with
ary structures when Al ions interact with them through Al to induce inhibitory or stimulatory effects (Exley and
oxygen-containing amino acids, side chains and protein Birchall, 1992). Interaction of Al with metabolic and other
backbone leading to ultimate denaturation (Mujika et al., enzymes causes inhibition or activation of the enzymes
2018) or conformational or structural alteration (Exley et (Hofstetter et al., 1987; Xu et al., 1990; Exley et al., 1994;
al., 1993; Zatta et al., 2005; Exley, 2006) as in β-amyloid. Zatta et al., 1999, 2000; Yang et al., 2003; Mailloux et al.,

Table 1. Toxic actions associated with aluminium exposure.


Toxic action or effect Selected references
Oxidative stress, lipid peroxidation Kattab et al., 2010; Exley, 2013; Abd-Elhady et al., 2013;
Zhang et al., 2016; Yang et al., 2018; Yu et al., 2019
Pro-inflammatory: organ inflammation in lung, intestine, heart, and testis Fogarty et al., 1998; Verma et al., 2007; Lerner, 2007; Exley,
2013; Taiwo, 2014; de Chambrun et al., 2014; Gherardi et al.,
2016; Martinez et al., 2017; Hangouche et al., 2017
Immunosupression: induces lymphocyte apoptosis and dysfunction, Nordal and Dahl, 1988; Kammalov et al., 2011; She et al., 2012; Zhu
inhibits lymphocyte proliferation, causes macrophage dysfunction et al., 2014; Zhuang et al., 2016; Xu et al., 2018; Yu et al., 2019
Protein denaturation and transformation Exley et al., 2006; Mujika et al., 2018

Enzymatic stimulation or inhibition Ohsaka and Nomura, 2016

Metabolic impairment: impairs glycolysis and Kreb’s Xu et al 1990; Mailloux et al., 2006
cycle; promotes lipid and protein oxidation
Genotoxicity: reduced cell proliferation and differentiation, dysneurogenesis Nam et al., 2014

Amyloidogenic and anti-amyloidolytic Sakamoto et al., 2006; Xu et al., 2016

Acts as metalloestrogen, promotes prolifera- Bakir and Darbre, 2015; Darbre, 2016
tion and migration of breast cancer cells
Induces teratogenesis causing foetal and neonatal defects Malekshah et al., 2005; Wang et al., 2012; El Mazoudy and Bekhet, 2016

Disrupts mineral metabolism of Fe, P, Ca, Zn, Cu by alter- Jeffery et al., 1996; Contini, 2007; Kell, 2009; Fu et al., 2014; Zhu et al., 2014
ing intestinal absorption and cellular uptake
Induces apoptosis, eryptosis, tissue necrosis Niemoeller et al., 2006; Xu et al., 2018; Yang et al., 2018; Yu et al., 2019

Disrupts cell membrane permeability and receptor function, Fu et al., 2014; Zhang et al., 2016; Sun et al., 2018; Gomes et al., 2019
increases osmotic fragility, inhibits membrane ATPases
Endocrine disruption: parathyroid hormone, testosterone, lutein- Díaz-Corte et al., 2001; Chinoy and Patel, 2001; Gonzelez-Suerez et al., 2005;
izing hormone, follicle stimulating hormone, estradiol, nor- Shahraki et al., 2008; Orihuela, 2011; Sun et al., 2011; Muselin et al., 2016;
epinephrine, cortisol, thyroid hormone, insulin Zhuang et al., 2016; Mouro et al., 2018; Wei et al., 2018 Gomes et al., 2019
Inhibits cartilage formation Zhang et al., 2017

Inhibits bone formation and mineralization by increasing osteo- Cox and Dunn, 2001; Li et al., 2012; Cao et al., 2016; Song et al., 2016; Sun et
clastic activity and reducing osteoblastic activity al., 2016; Yang et al., 2016; Huang et al., 2017; Yang et al., 2018; Xu et al., 2018
Induces hypertension (systolic and arterial) Zhang et al., 2016

Causes ischaemic stroke and thrombosis Abedini et al., 2014

Induces contact allergy Netterlid et al., 2013

Inhibits the biological function of vitamin D in the Dunn et al., 1995


intestine linked to calcium absorption

ISSN: 1337-6853 (print version) | 1337-9569 (electronic version)


Interdisciplinary Toxicology. 2019; Vol. 12(2): 45–70 51
Full-text also available online on PubMed Central

Figure 4. Cellular pathology in the systemic toxicosis of aluminium.

Figure 5. Metabolic abnormalities in aluminium toxicosis.

2006; Sushma et al., 2007; Ohsaka and Nomura, 2016). Al et al., 2012; Xu et al., 2018; Yang et al., 2018; Yu et al.,
binds to the phosphate groups of nucleotide such as ade- 2019). The oxidative injury was reported to activate the
nosine triphosphate (ATP) and affects energy metabolism JNK apoptotic pathway in ostoblasts (Yang et al., 2018).
(Kawahara et al., 2007). Exposure of hepatocytes to Al In culture, Al induced apoptosis of osteoblasts by inhibit-
impedes ATP production, inhibits glycolysis, impairs the ing apoptotic Bcl-2 protein expression and increasing the
function of tricarboxylic acid (Kreb’s) cycle and promotes expression of pro-apoptotic Bax, Bak and Bim proteins
lipid and protein oxidation (Xu et al., 1990; Mailloux et al., (Xu et al., 2018). Al may decrease ferritin synthesis and
2006) with a metabolic shift to lipogenesis in tissues (Han increase the expression of transferrin receptors, thereby
et al., 2013). These metabolic perturbations (Figure 5) disrupting the normal synthesis of transferrin receptors
may be responsible for the reports of body weight loss and with ferritin creating increased free iron levels in the
decreased production performance (like egg production) cell, resulting in an increase of oxidative damage via the
in animals exposed to Al (Wisser et al., 1990; Capdevielle fenton reaction (Yamanaka et al., 1999). The activities of
and Scanes, 1995; Li et al., 2015). superoxide dismutase (SOD), catalase (CAT), glutathione
Al exposure can cause the disruption of iron homeo- peroxidase (GPx) and glutathione (GSH) are affected
stasis leading to iron overload (Ward et al., 2001; Contini by Al exposure because of oxidative stress (Oteiza et
et al., 2007). Oxidative stress and injury, mediated by iron, al., 1993a; Julka and Gill, 1996; Campbell et al., 1999).
seems to be facilitated by Al (Xiea et al., 1996). Elevated Abnormal increases in levels of malondealdehyde (MDA)
concentrations of cellular iron can enhance oxidative and thiobarbituric acid reactive substances (TBARS) were
damage to the cell and are linked to the pathogenesis reported along with decreased levels of antioxidants such
of neurodegenerative disorders (Jang and Surh, 2002; as GSH, GPx, SOD, and CAT in tissue homogenates of rats
Toyokuni, 2002; Adzersen et al., 2003; Deugnier, 2003; exposed to Al (Anane and Creppy, 2001; Gonzalez et al.,
Ng, 2004). Iron overload due to Al exposure has been 2007; Newairy et al., 2009; Khattab et al., 2010; Bai et al.,
shown to result in increased lipid peroxidation, DNA 2012; Exley, 2013; Abd-Elhady et al., 2013; Zhang et al.,
lesions, and apoptosis induced by reactive oxygen species 2016; Yu et al., 2019).
(Bacon et al., 1983; Oteiza, 1994; Kell, 2009). Apoptosis Mutagenesis and alteration of gene function may arise
of erythrocytes (eryptosis), lymphocytes and osteoblasts from the toxic action of Al with changes in transcrip-
is also stimulated by Al ions (Niemoeller et al., 2006; Li tional expressions (Exley, 2013). Somatic and germinal

Copyright © 2019 SETOX & Institute of Experimental Pharmacology and Toxicology, CEM SASc.
52 Aluminium toxicosis
Ikechukwu Onyebuchi Igbokwe, Ephraim Igwenagu, Nanacha Afifi Igbokwe

genetoxicity in mice exposed to Al was associated with secretion of matrix metalloproteinase (MMP9) and levels
chromosomal aberrations and depression of mitosis of activated MMP14 that are involved in migratory and
(D’Souza et al., 2014). Neuronal gene expression is influ- invasive properties of cancerous cells, thereby influencing
enced by the binding of Al to DNA (Lukiw et al., 1998) the metastatic process (Darbre et al., 2013a, b; Bakir and
predisposing cells to or causing genotoxicity (Pogue and Darbre, 2015; Darbre, 2016). It is unclear whether Al has
Lukiw, 2016) and thus Al exposure may lead to reduction the capacity to initiate and promote any other carcino-
of cell proliferation and differentiation (Nam et al., 2014, genic process apart from the indirect evidence provided
2016; Sun et al., 2015; Cao et al., 2016; Li et al., 2016; Yang above with regard to breast cancer.
et al., 2016; Sun et al., 2016, 2017; Huang et al., 2017). Pro-inflammatory actions of Al have been reported in
Neurogenesis was impaired by Al toxicity (Nam et al., various tissues (Fogarty et al., 1998; Verma et al., 2007;
2014, 2016). Osteoblastic proliferation and differentiation Lerner, 2007; Exley, 2013; Taiwo, 2014; de Chambrun
were inhibited by Al when there was downregulation and et al., 2014; Gherardi et al., 2016; Martinez et al., 2017;
inhibition of Wnt/β-catenin signaling pathway (Sun et Hangouche et al., 2017). It is triggered by Al-induced
al., 2015; Cao et al., 2016; Huang et al., 2017; Sun et al., oxidative stress and free radical production (Milnerowicz
2017). Osteoblast differentiation was also inhibited by et al., 2015). Exposure to Al increased pro-inflammatory
Al through the inhibition of BMP-2 signaling pathway cytokine (interleukin-1β and tumor necrosis factor alpha)
(Yang et al., 2016). In addition, osteoblast mineralization levels (Jangra et al., 2015) and elevated gene expression of
in vitro was inhibited by Al-induced decline in trans- tumor necrosis factor alpha (TNFalpha) and macrophage
forming growth factor (TGF)-β1 expression and action, inflammatory protein-1alpha (MIP-1alpha)  in concen-
and upregulation of Smad7 expression (Sun et al., 2016) tration-dependent manner (Johnson and Sharma, 2003).
along with decreased protein expressions of osteopontin, Genes that encode pro-inflammatory signaling elements
osteocalcin and osteosialoprotein (Song et al., 2017). The were significantly up-regulated by Al (Lukiw et al., 2005).
mineralization of bone is impaired by decreased calcium These cytokines that are released due to Al exposure can
absorption (Orihuela, 2007), because Al inhibits the syn- recruit leukocytes, which secrete more pro-inflammatory
thesis of calbindin, a calcium-binding protein involved cytokines and other chemokines, to exacerbate the
in transcellular transport of calcium in enterocytes and inflammation (Milnerowicz et al., 2015). The inflamma-
inhibits the stimulation of synthesis of osteocalcin (the tion can be a chronic granulomatous type (Chen et al.,
bone matrix protein) in osteoblasts by vitamin D via cel- 1978; de Vuyst et al., 1987; Forgarty et al., 1998; Gherardi
lular unresponsiveness (Fanti et al., 1992; Jeffery et al., and Authier, 2012) and Al has been reported to cause
1996; Cox and Dunn, 2001). The expression of cartilage granuloma formation in vitro (de Chambrun et al., 2014).
stimulating growth factors, TGF-β1 and BMP-2, were Chronic exposure of mice (5 months) to Al sulfate in
inhibited by Al, thereby suppressing cartilage growth drinking water elicited time-dependent systemic inflam-
and disrupting cartilage structure (Zhang et al., 2017). mation characterized by increased serum  interleukin-6
The effects of Al on growth manifested in developmental (IL-6) and tumor necrosis factor alpha (TNFα), C-reactive
abnormalities of fetuses due to teratogenesis in pregnant protein (CRP) and a triad of pro-inflammatory microR-
individuals (Malekshah et al., 2005; Wang et al., 2012; El NAs (miRNA-9, miRNA-125b and miRNA-146a) and
Mazoudy and Bekhet, 2016; Yassa et al., 2017). the biomarkers of inflammation   indicated progressive
The proliferative and migratory characteristics of the chronic inflammation in the exposed animals (Pogue et
human breast cancer cell may be affected by Al when it al., 2017). The inflammatory conditions associated with
acts as a metalloestrogen or increases the intracellular Al exposure are summarized in Figure 6.

Figure 6. Inflammatory conditions in aluminium toxicosis.

ISSN: 1337-6853 (print version) | 1337-9569 (electronic version)


Interdisciplinary Toxicology. 2019; Vol. 12(2): 45–70 53
Full-text also available online on PubMed Central

Table 2. Effects of aluminium exposure on endocrine secretions in animals.


Hormone Animal/human Increase Decrease Normal Reference
Plasma growth hormone Duckling + Capdevielle et al., 1995
Plasma insulin-like growth Duckling + Capdevielle et al., 1995
factor 1
Plasma cortisol Rat + Vasanthan and Joshi, 2018
Blood norepinephrine Rat + Zhuang et al., 2016
Serum estradiol Mice + Chinoy and Patel, 2001
Serum testosterone Gerbil + Reza and Palan, 2006; Gomes et al., 2019
(Meriones unguiculatus)
Rat + Shahraki et al., 2008; Sun et al., 2011; Muselin et al., 2016;
Mouro et al., 2018
Serum luteinizing hormone Rat + Shahraki et al., 2008; Sun et al., 2011; Muselin et al., 2016
Rat + Reza and Palan, 2006
Serum follicule stimulating Rat + Reza and Palan, 2006; Sun et al., 2011
hormone Rat + Shahraki et al., 2008
Serum/plasma parathyroid Rat + Cannata et al., 1983; Díaz-Corte et al., 2001; Gonzelez-Suerez
hormone et al., 2005
Human patient with + Sherrard et al., 1985; Cournot-Witmer and Plachott, 1990
chronic renal failure, on
haemodialysis
Serum thyroid hormone, T4 Rat + Orihuela, 2011
Serum thyroid hormone, T3 Rat + Orihuela, 2011
Serum thyroid hormone, Rat + Orihuela, 2011
free T4
Serum thyrotropin (TSH) Rat + Orihuela, 2011
Insulin Rat + (Acute) + (Chronic) Wei et al., 2018

As an adjuvant, Al in vaccines induces local inflamma- to the glands through oxidative stress, thereby decreas-
tion that involves the NLRP3 inflammasome and NLRP3- ing the level of the hormones secreted (Morrissey et al.,
independent pathways where macrophages, B- and 1983) into the bloodstream for action at the target organs,
T-lymphocytes play important roles in enhancing antigen- causing organ hypofunction. For instance, there are
specific immune responses and increasing inflammatory reports of testicular and ovarian failures (Mohammed
cytokine production (Exley et al., 2010; Hogenesch, 2013; et al., 2008; Fu et al., 2014; Miska-Scramm et al., 2017)
Zlatkovic et al., 2013; He et al., 2015). Toxic exposure to Al from inadequate androgenic hormone levels (Chinoy and
causes immunotoxicity leading to inhibition of lympho- Patel, 2001; Shahraki et al., 2008; Sun et al., 2011; Muselin
cyte and macrophage functions (Nordal et al., 1988; Zhu et al., 2016; Mouro et al., 2018) and decreased androgen
et al., 2014a). Immunosuppression arises from oxidative receptor functions (Fu et al., 2014; Sun et al., 2018;
stress which is associated with apoptosis of lymphocytes Gomes et al., 2019), bone pathology due to dysfunction
(Yu et al., 2019) and damage to thymocytes and lympho- of parathyroid gland (Cannata et al., 1983; Sherrard et al.,
cytes (Kamalov et al., 2011). Immune functions of splenic 1985; Cournot-Witmer and Plachott, 1990; Díaz-Corte et
B- and T-lymphocytes were inhibited in vitro by reduc- al., 2001; Gonzelez-Suerez et al., 2005), prediabetes and
tion of lymphocyte proliferation, cytokine secretion and diabetes due to pancreatic islet damage (Wei et al., 2018).
proportions of CD-3(+) and CD-4(+) lymphocytes (She et Parathyroid function can be impaired by the Al ion act-
al., 2012). LPS-induced NLRP3 inflammasome activation, ing on calcium-sensing receptors when calcium level is
IL-1β, IL-6 and TNF-α expression and release in perito- low, with a higher efficiency than calcium and decreasing
neal macrophages were also suppressed by Al exposure the expression of the receptors in the gland (Gonzelez-
(Xu et al., 2018). Norepinerphrine release and activation Suerez et al., 2005). The metabolic effect of some levels of
of β-adenoceotors/cAMP pathway were promoted by Al thyroxine (T3 and T4) decline without change in free T4
in vivo, and this endocrine factor suppressed macrophage level is yet to be ascertained (Orihuela, 2011). The secre-
expressions of MIF and TNF-α (Zhuang et al., 2016). tion of the hormone, sometimes increases when Al ion is
Contact allergy to Al has been reported as an aberrant stimulatory to the gland or because the target organs are
immune response in those having atopic dermatitis refractory or unresponsive to the hormone when there is
(Netterlid et al., 2013). receptor depletion or decreased expression of the recep-
The endocrine disruptions or hormonal changes tor on the cell membrane. The Al ions act as chemical
associated with Al exposure are summarized in Table 2. stressor by promoting the release of norepinerphrine
Al accumulates in endocrine glands and causes damage (Zhuang et al., 2016) and cortisol (Vasanthan and Joshi,

Copyright © 2019 SETOX & Institute of Experimental Pharmacology and Toxicology, CEM SASc.
54 Aluminium toxicosis
Ikechukwu Onyebuchi Igbokwe, Ephraim Igwenagu, Nanacha Afifi Igbokwe

2018); and the elevated levels of these hormones can cause al., 2018), inaction of membrane-bound enzymes, inhibi-
increase in blood pressure (Zhang et al., 2016). Elevated tion of calbindin protein level in enterocytic membrane
blood insulin level is associated with insulin resistance (Cox and Dunn, 2001) and downregulation of GLUT4
due to depletion of glucose transporter-4 protein expres- protein expression in the membrane of skeletal muscle
sion in skeletal muscles during Al exposure (Wei et al., (Wei et al., 2018). The surface of cell membranes could
2018). Pseudohyperparathyroidism is associated with be affected by Al exposure through externalization of
osteitis fibrosa in human patients with renal failure and phosphatidylserine after apoptosis (Vota et al., 2012),
hypercalcemia when exposed to Al intoxication (Sherrard inhibition of membrane receptor protein expression in
et al., 1985). gonads (Fu et al., 2014; Sun et al., 2018; Gomes et al.,
The cellular membrane is vital for the viability of 2019), dysregulation of erythropoietin receptor func-
the cell and Al exposure disrupts membrane activity tions on erythroid progenitors (Vittori et al., 2005) and
via oxidative stress in various ways. In Alzheimer’s loss of membrane surface sialic acid residues on vascular
disease associated with Al exposure, membrane fluidity endothelial tissue with impaired intercellular junctions
increased in platelets and decreased in erythrocytes; (Vorbrodt et al., 1994).
and this observation was corroborated by a study
where in vitro exposure of membrane suspensions to Al
increased fluidity of platelet membranes and decreased Systemic toxicosis
the fluidity of erythrocyte membranes (van Rensburg
et al., 1992) with consequent effect on the viability of Pulmonary effect
platelets (Neiva et al., 1997) and erythrocytes (Vittori Pulmonary lesions in humans linked to Al exposure
et al., 2002). Erythrocyte membrane permeability and during production of Al products include granulomatous
osmotic fragility are affected by in vivo and in vitro Al pneumonia, pulmonary granulomatosis, pulmonary
exposures (Igbokwe, 2018). Erythrocyte osmotic fragil- fibrosis, pulmonary alveolar proteinosis and desquama-
ity decreased (Bazzoni et al., 2005) or increased (Zatta tive interstitial pneumonia (Chen et al., 1978; Herbert et
et al., 1989; Hernández et al., 2008; Al-Qayim et al., al., 1982; Miller et al., 1984; De Vuyst et al., 1987; Jederlinic
2014; Oztürk and Ozdemir, 2015, Zhang et al., 2016; et al., 1990; Taiwo, 2014; Iijima et al., 2017). Asthma may
Cheng et al., 2018) depending on the Al speciation and be caused by Al exposure (Burge et al., 2000), though the
type of erythrocyte injury. Eryptotic (apoptotic) injury asthma among Al workers may be due to other chemical
reduces the erythrocyte aggregate size (Bazzoni et al., factors like gases and smoke (Taiwo et al., 2006). Reactive
2005) because of the shrinking effect, thereby increasing airways dysfunction syndrome was rarely reported
osmotic resistance (Igbokwe, 2016). On the other hand, among Al smelter workers (Wesdock and Arnold, 2014).
eryptotic injury which progresses to oncotic injury may Acute-duration oral exposure to Al phosphide has been
cause swelling of the erythrocyte and increase osmotic reported to cause pulmonary edema in persons following
fragility. It is therefore presumed that Al may cause either accidental or volitional ingestion (Chopra et al., 1986;
shrinking or swelling effect when the erythrocyte mem- Khosla et al., 1988). The toxicity was probably due to the
brane is destabilized by Al exposure because of altered formation of highly toxic phosphine gas rather than to
membrane permeability to intracellular and extracellular Al exposure (Alter et al., 2001; Kamanyire and Murray,
ions (Igbokwe, 2016). Cell membrane functions which 2003; Moghadamnia, 2012). Intermediate- and chronic-
regulate transmembrane transport of ions are expected duration studies found no organ weight or histological
to be disrupted when ATPase in cell membrane loses changes in the lungs of rats exposed to 70 mg Al/kg/day as
some level of activity during Al exposure. There are Al chloride in drinking water for 30, 60 or 90 days (Dixon
reports showing that Al inhibited activities of Na+K+- et al., 1979), rats exposed to 133 mg Al/kg/day as Al nitrate
ATPase, Mg2+-ATPase and Ca 2+-ATPase in erythrocytes in drinking water for 30 days (Gomez et al., 1986), rats and
(Zhang et al., 2016), vascular endothelial cells (Vorbrodt mice exposed to 0.6 or 1.2 mg Al/kg/day as Al potassium
et al., 1994), testes (Sun et al., 2018) and ovaries (Fu et al., sulfate in drinking water for 24 months (Schroeder and
2014) of rats. The Al-induced change in erythrocyte size Mitchener, 1975a, b), or mice exposed to 979 mg Al/kg/
may also be accompanied by change in erythrocyte shape day as Al potassium sulfate in food for 20 months (Oneda
resulting in the formation of echinocytes (Suwalsky et et al., 1994). However, Hasseeb et al. (2011) reported
al., 2004), acanthocytes and stomatocytes (Vittori et al., neutrophilic and mononuclear cell infiltrations of lung
2002) in vitro, due to altered membrane morphology alveoli of rats administered 37 mg/kg/day of Al chloride
(Lukyanenko et al., 2013). After long-term oral intake of in drinking water for 8 weeks. Congested blood vessels in
Al, schistocytes and target cells were observed in stained inter-alveolar spaces were reported after administration
peripheral blood of rats (Vittori et al., 1999). The lipid of different concentrations of Al chloride via gavage for 8
bilayer of the plasma and mitochondrial membranes was weeks (Buraimoh and Ojo, 2013). Pulmonary lesions are
morphologically altered in lymphocytes (Skarabahatava rare and inconsistent in experimental animals where Al
et al., 2015). The protein components of membranes are exposure is not through aerosol vehicles. Under natural
degraded or inadequately expressed during Al exposure, conditions, the vehicular substances and the Al specia-
as observed in the loss of band 3 protein of erythrocyte tion may influence the stimulation of chronic pathologic
membrane (Vittori et al., 2002; Vota et al., 2012; Cheng et reactions in the lung.

ISSN: 1337-6853 (print version) | 1337-9569 (electronic version)


Interdisciplinary Toxicology. 2019; Vol. 12(2): 45–70 55
Full-text also available online on PubMed Central

Cardiovascular effects Al and lipopolysaccharides (de Chambrun et al., 2014).


Toxic myocarditis, myocardial hypokinesia, left ventricu- Oral Al chloride exposure caused epithelial degeneration,
lar thrombosis and myocardial dysfunction were reported goblet cell proliferation and lymphocyte infiltration in the
in a case of Al phosphide intoxication (Hangouche et al., mucosa of the small intestine of Wistar rats (Buraimoh
2017). Ischemic stroke due to thrombosis in the right and Ojo, 2012). Few experimental studies (Gomez et al.,
middle cerebral artery was reported as the delayed 1986; Oneda et al., 1994) did not report intestinal lesions
complication of Al phosphide poisoning (Abedini et al., after oral exposure to Al at 133 mg Al/kg/day as Al nitrate
2014). However, other Al compounds may not cause car- in drinking water to rats for 30 days and 979 mg Al/kg/day
diovascular lesion. Cardiac teratogenessis was reported as Al potassium sulfate in the food of mice for 20 months.
in embryonic chick heart where defects in ventricular The acute and chronic inflammations in the intestine
septation and ventricular myocardium were reported may induce poor intestinal digestion and absorption.
(El Mazoudy and Bekhet, 2016). There was significant
association between increased maternal hair Al contents Hematologic effects
and risk of total congenital heart defects in offspring, Al exposure has been associated with significant inhibi-
especially in subtypes such as septal defects, conotrun- tion of colony forming units-erythroid (CFU-E) develop-
cal defects and right ventricular outflow obstruction in ment in the bone marrow of mice exposed to 13 mg Al/
female rats (Wang et al., 2012). No histological changes kg as Al citrate or chloride administered via gavage for
were observed in the hearts of rats given 70 mg Al/kg/ 5 days/week for 22 weeks (Garbossa et al., 1996), rats
day as Al chloride in drinking water for 30, 60, or 90 days exposed to 27 mg Al/kg as Al citrate administered via
(Dixon et al., 1979). Similarly, no effect on organ weight gavage 5 days/week for 15 weeks (Garbossa et al., 1998),
nor histological changes were found in the hearts of rats and rats exposed to 230 mg Al/kg/day as Al citrate in
that ingested 133 or 284 mg Al/kg/day as Al nitrate in drinking water for 8 months (Vittori et al., 1999). The
drinking water or base diet for 30 days (Gomez et al., effect of Al on erythroid progenitor cells and erythrocytes
1986) or 100 days, respectively (Domingo et al., 1987). was associated with slow growth and increased degrada-
Organ weight and histological changes were not observed tion of membrane band 3 proteins, respectively (Vittori
in the hearts of dogs that consumed 75 mg Al/kg/day et al., 2002). The genotoxicity from Al exposure in mice
(Katz et al., 1984) or 88 mg Al/kg/day (Pettersen et al., resulted in mitodepressive effect in the bone marrow
1990) as sodium Al phosphate in the diet for 6 months. (D’Souza et al., 2014). Anemia caused by Al toxicity is
In summary, cardiovascular effects due to toxicosis are not associated with adequate regenerative activity of
congenital heart defects, inflammation and dysfunction the bone marrow and reticulocytosis (Chmielnicka et
of the myocardium and cardiovascular thrombosis. al., 1994; Osman et al., 2012). The additional causes of
anemia appear to be multi-factorial and include defective
Gastrointestinal effects hemoglobin production due to inhibition of the enzymes
In horses, Al was found in tissues, blood vessel walls and of heme synthesis, altered erythrocyte membrane struc-
granulomatous lesions of the intestines associated with ture and fragility, shortening of red blood cell life span
equine granulomatous enteritis (Fogarty et al., 1998), due to eryptotic and oncotic injuries, and inadequate iron
and Al was demonstrated to have the capacity to induce utilization (Zatta et al., 1989; Perez et al., 2001; Bazzoni
granuloma formation in vitro (de Chambrun et al., 2014). et al., 2005; Vittori et al., 2002; Niemoeller et al., 2006;
Oral intake of Al may affect the intestinal microbiota, Hernández et al., 2008; Sadhana, 2011; Vota et al., 2012;
permeability and immune response which influence the Lukyanenko et al., 2013; Al-Qayim et al., 2014; Oztürk
local inflammatory conditions (Vignal et al., 2016). In and Ozdemir, 2015; Zhang et al., 2016; Cheng et al.,
individuals that are genetically susceptible to Crohn’s dis- 2018). Significant decreases in hemoglobin, hematocrit
ease, Al is linked to the induction and persistence of the (packed cell volume) and erythrocyte osmotic fragility
chronic relapsing intestinal inflammation (Lerner, 2007). were reported after Al exposure (Garbossa et al., 1996;
Inflammatory bowl diseases, consisting of disease entities Garbossa et al., 1998; Vittori et al., 1999; Farina et al.,
like Crohn’s disease and ulcerative colitis, are character- 2005). The anemia is characterized by decreases in mean
ized by excessive intestinal inflammation and experimen- corpuscular volume (microcytosis) and mean corpuscular
tal evidence in mice indicates that Al promotes intestinal hemoglobin (hypochromia), but in chronic exposures,
inflammation, thereby implicating Al in the pathogenesis the erythrocyte parameters recover with persistence of
of inflammatory bowl diseases (de Chambrun et al., 2014). microcytosis and hypochromia (Mahieu et al., 2000). In
Chemically-induced acute colitis and chronic colitis in rats loaded with Al, heme dyshomeostasis was reported
transgenic mice lacking interleukin 10 were aggravated with evidence of decreased activity of aminolevulinic acid
by oral exposure to Al, because Al increased the intensity dehydratase and increased activity of heme oxygenase in
and duration of intestinal inflammation and decreased the rat liver associated with activation of JNK pathway,
regeneration or renewal of the intestinal epithelial indicating an increase in heme degradation (Lin et al.,
mucosal cells (de Chambrun et al., 2014). Furthermore, 2013). No alterations in hemoglobin, hematocrit and
intestinal barrier function was impaired by Al exposure erythrocyte osmotic fragility were reported in a number
under basal conditions; and there was a synergistic of experimental Al exposures (Katz et al., 1984; Gomez
stimulation of pro-inflammatory cytokine expression by et al., 1986; Domingo et al., 1987; Pettersen et al., 1990;

Copyright © 2019 SETOX & Institute of Experimental Pharmacology and Toxicology, CEM SASc.
56 Aluminium toxicosis
Ikechukwu Onyebuchi Igbokwe, Ephraim Igwenagu, Nanacha Afifi Igbokwe

Table 3. Summary of haematologic effects of aluminium toxicosis.


kg/day) for 90 days accumulated more Al in their brains,
had increased brain acetyl cholinesterase activity and had
Toxic effects Toxic actions
decreased brain choline acetyltransferase activity (Bilkei-
Depressed Inhibition of CFU-E Gorzó, 1993). Mice fed high Al levels (1,000 mg/kg diet
erythropoiesis
Slow growth of erythroid cells of Al as Al lactate) were less active, had decreased grip
Inhibition of heme synthesis strength, and increased startle responses after 90 days
when compared with control (Golub et al., 1992). Oteiza
Increased heme degradation
et al. (1993b) reported that mice fed diets containing
Dysregulated erythropoietin receptor function
1,000 mg/kg diet of Al (as Al chloride) with sodium citrate
Anaemia Reduced erythrocyte life span accumulated more Al in the brain nuclear fraction and
Erythrocyte apoptosis (eryptosis) spinal cord, had lower grip strength, and greater startle
Altered erythrocyte fragility responsiveness after 5 and 7 weeks. Old (18 months of
Decreased erythrocyte membrane fluidity
age) rats exposed to Al (100 mg/kg/day) in drinking water
with citrate (356 mg/kg/day of citrate) had decreased
Inhibition of erythrocyte membrane ATPase
numbers of synapses and a greater percentage of perfo-
Altered erythrocyte shape: echinocytes, rated synapses than controls, but no changes in behavior
acanthocytes, stomatocytes, target cells
(Colomina et al., 2002). Garruto et al. (1989) reported that
cynomolgus monkeys fed a low calcium diet (3,200 mg/
kg diet) with Al (125 mg/day) for 41 to 46 months had
Oteiza et al., 1993b; Garbossa et al., 1996). Vittori et more degenerative changes that were consistent with
al. (1999) did not find significant alterations in plasma early Alzheimer’s disease or Parkinson’s dementia in the
iron levels or total iron binding capacity in rats exposed central nervous system than control monkeys. Golub and
to 230 mg Al/kg/day as Al citrate in drinking water for Germann (2001) observed growth depression and poorer
8 months; however, they reported impaired iron uptake performance on standardized motor tests in mice off-
and decreased iron incorporation into heme in the bone spring when dams were exposed to Al (1,000 mg/kg diet as
marrow. Farina et al. (2005) found significant decreases Al lactate) with marginal levels of calcium and magnesium
in blood iron concentrations and no change in total iron during pregnancy and lactation. Mice fed lower rather
binding capacity in rats exposed to 54.7 mg Al/kg/day as than recommended levels of calcium (2,500 versus 5,000
Al sulfate in a sodium citrate solution in drinking water mg/kg diet of calcium) with Al (15,600 mg/kg diet as Al
for 18 months. Florence et al. (1994) reported decreases hydroxide) for 11 to 25 months accumulated more hyper-
in serum iron levels, total iron binding capacity, and phosphorylated tau protein in the cortical neurons and
transferrin saturation in rats exposed to 75 mg Al/kg/day had more atrophic neurons in the central nervous system
as Al citrate in the diet for 6 months. Chronic Al exposure (Kihira et al,. 2002). Transgenic mice with over-expressed
in rats disrupted iron homeostasis (Zhang et al., 2010). In human amyloid precursor protein had increased brain
summary, the hematologic effect of toxicosis consists of isoprostane levels and more amyloid-β peptide formation
anemia due to erythrocyte and erythroid pathology with and deposition when Al was added to their diets, but the
suppression of erythropoiesis (Table 3) effects of Al were reversed by additional dietary vitamin E
(Pratico et al., 2002), suggesting that Al could contribute
Neurologic effects to neurodegeneration by enhancing amyloid deposition
In humans, Al accumulation in the brain and scalp hairs and aggravating lesions by oxidative events (Campbell and
has been associated with neurodegenerative diseases such Bondy, 2000; Yuan et al., 2012; Chen and Zhong, 2014;
as dialysis-associated encephalopathy, Alzheimer’s dis- Liaquat et al., 2019). In a nutshell, Al exposure promotes
ease, Parkinson’s disease (dementia), amyotropic lateral oxidative stress and amyloid deposition in the nervous tis-
sclerosis, multiple sclerosis and autism (King et al., 1981; sue which results in neurodegeneration, neuronal necrosis
Savory et al., 1996; Kawahara and Kato-Negishi, 2011; and dysneurogenesis, which constitute the basis for the
Arain et al., 2015; Jones et al., 2017; Mirza et al., 2017; neurological diseases associated with Al intoxication.
Mold et al., 2018). The Al in brains of 5 out of 12 donors
with familial Alzheimer’s disease was > 10 µg/g dry weight Musculoskeletal effects
(Mirza et al., 2017). In autism, Al in parts of the brain was The major myopathy induced by Al exposure is macro-
up to 19 µg/g dry weight (Mold et al., 2018). There is a role phagic myofasciitis (aluminic granuloma) associated
for Al in multiple sclerosis because patients excrete high with chronic arthromyalgia or myalgia and chronic
amounts of Al in urine, facilitated by drinking silicon-rich fatigue syndrome (Exley et al., 2009; Gherardi and
mineral water (Jones et al., 2017). Subchronic exposure Authier, 2012; Rigolet et al., 2014; Gherardi et al., 2016;
to Al was associated with reduced population of neural Miller, 2016). Skeletal muscle necrosis occurred in the
stem cells and hampered cell proliferation and neuroblast diaphragm and abdominal muscles of rats adjacent to the
differentiation in the brain of mice (Nam et al., 2014, peritoneum after intraperitoneal injection of Al lactate
2016). Injection of Al, especially intra-cisternally, induced (Levine et al., 1992). Muscle fiber atrophy, with retarda-
neurological changes in animal models (Wisniewski et al., tion of growth, was reported in growing pigs which was
1980; Anon, 2008c). Rats orally administered Al (100 mg/ associated with hypophosphatemia induced by dietary Al

ISSN: 1337-6853 (print version) | 1337-9569 (electronic version)


Interdisciplinary Toxicology. 2019; Vol. 12(2): 45–70 57
Full-text also available online on PubMed Central

hydroxide supplementation (Haglin et al., 1994). Smooth administered Al-containing phosphate-binding agents
muscle contraction induced by K+ ion was inhibited by Al (Mayor et al., 1985; Wills and Savory, 1989; Andreoli,
exposure (Nasu et al., 1998). Myocardial function may be 1990). Low osseous remodeling rate and peripheral
altered in diabetic individuals by Al exposure, in as much resistance to parathyroid hormone are associated with
as Al toxicity potentiates the decline in calcium uptake Al intoxication (Pun et al., 1990). Decreased Al urinary
into the sarcoplasmic reticulum of the myocardial fibers excretion caused by impaired renal function with, possi-
of such individuals (Levine et al., 1990). In individuals bly, an increase in gastrointestinal absorption of Al results
where neurodegerative conditions affect the nerve supply in increased Al load leading to markedly increased bone
to muscles, the muscles may undergo denervation atro- Al levels and the presence of Al between the junction of
phy and become dysfunctional as in multiple sclerosis or calcified and non-calcified bones (Alfrey, 1993). Long-
amyotropic lateral sclerosis. Taken together, Al toxicosis term oral exposure to Al results in an increase in Al levels
may cause muscle damage, inflammation and dysfunction in the bone (Ahn et al., 1995; Konishi et al., 1996) that is
The bone diseases associated with Al exposure are responsible for the bone disease.
osteoporosis, osteomalcia, rickets, exostosis, osteodystro- In brief, the review has identified the following events
phy and osteitis fibrosa (Sherrard et al., 1985; Chappard et to occur during Al exposure to disrupt bone morphol-
al., 2016; Rodríguez and Mandalunis, 2018; Klein, 2019). ogy: (a) interference with the availability of calcium for
There is increased risk of osteoporosis and low bone min- bone formation at the level of intestinal absorption and
eral density during Al exposure (Cao et al., 2016; Sun et al., hormonal control through parathyroid hormone; (b) inhi-
2016) because of disruption of bone formation, and inhibi- bition of osteoid formation and mineralization through
tion of osteoblast proliferation, differentiation and miner- osteoblast dysregulation; and (c) destabilization of osteo-
alization (Li et al., 2012, 2016; Cao et al., 2016; Sun et al., clast functions with alteration in osteoclastogenesis and
2016; Yang et al., 2016; Zhu et al., 2016b; Song et al., 2017; osteoclast apoptosis (Figure 7).
Sun et al., 2017; Huang et al., 2017). In individuals with Al
overload, undecalcified bone matrix contains Al and bone Reproductive and developmental effects
conditions like exostosis and osteomalacia may occur in Human reproduction may be affected negatively by Al
circumstances that increase Al uptake and colocalization exposure (Klein et al., 2014; Mouro et al., 2017). Human
as observed in celiac disease, hemochromatosis and sickle semen and spermatozoa contain Al and patients with
cell anemia (Chappard et al., 2016). Osteoclastogenesis is oligospermia had higher Al concentration than healthy
promoted by low-dose exposure while osteoclast apoptosis individuals (Klein et al., 2014). At human dietary level
is caused by high-dose exposure (Yang et al., 2018). There of Al and continuous exposure for 60 days, the rat testes
are case reports of osteomalacia and rickets in infants accumulated low Al levels of 3.35 µg/g and it was associ-
and adults using Al-containing antacids for the treat- ated with increased oxidative stress and inflammation,
ment of gastrointestinal illnesses (Chines and Pacifici, decreased daily sperm production, reduced sperm count
1990; Pivnick et al., 1995; Woodson, 1998). The Al in and motility and increase in abnormal spermatozoa
antacids binds with dietary phosphorus and prevents its (Martinez et al., 2017). In male rats, subchronic exposure
absorption resulting in hypophosphatemia and phosphate to Al chloride did not result in elevated Al accumula-
depletion (Woodson, 1998). Osteomalacia, characterized tion in the testes, but toxic effects reported in the testes
by bone softening, increased spontaneous fractures and included impairment of spermatogenesis and increase in
pain, has been reported in dialyzed uremic adults and sperm malformation rate (Zhu et al., 2014b). Imbalance
children exposed to Al-contaminated dialysate or orally in trace mineral metabolism occurred in the testis with

Figure 7. Pathogenesis of bone disease in aluminium toxicosis.

Copyright © 2019 SETOX & Institute of Experimental Pharmacology and Toxicology, CEM SASc.
58 Aluminium toxicosis
Ikechukwu Onyebuchi Igbokwe, Ephraim Igwenagu, Nanacha Afifi Igbokwe

testicular levels of iron and zinc increasing and that of D’Souza et al., 2014). Testicular and epididymal weights
copper decreasing during exposure (Zhu et al., 2014b). and serum testosterone and luteinizing hormone levels
Furthermore, metabolic inhibition in the testis was were reduced by Al exposure (Reza and Palan, 2006;
reported with regard to the functions of acid phospha- Mouro et al., 2017). In male and female gerbils (Meriones
tase, succinate dehydrogenase, and lactate dehydroge- unguiculatus), Al exposure disrupted prostate develop-
nase and its isoenzymes (Zhu et al., 2014b), alongside ment in neonates, with the consequence of adult off-
with testicular membrane dysfunction due to inhibition spring having elevated serum testosterone levels with low
of membrane ATPase activities in Al-exposed rats (Sun androgen receptor frequency associated with increased
et al., 2018). The weights of the testes and epididymides proliferation of cells of the prostate (Gomes et al., 2019).
were decreased by Al exposure in rats as serum testos- Chinoy and Patel (2001) exposed female mice to Al
terone levels dropped (Mouro et al., 2018). In male rats, chloride at 200 mg/kg for 30 days and observed decreased
testicular development was impaired by Al exposure, steroidogenesis in the ovaries associated with decreased
associated with reduction in serum levels of testosterone serum estradiol levels. Exposure to Al sulphate during
and luteinizing hormone (LH) levels and decrease in gestation caused reduction in maternal body weight,
androgen receptor protein expression without effect on reduction in fetal weight and crown rump length, and
serum follicle stimulating hormone (FSH) (Sun et al., impairment of fetal bone development and preossifica-
2011, 2018). The offspring of exposed rats (F0) in a three- tion (Yassa et al., 2017). In adult mice exposed to Al at
generation study belonging to F1 and F2 had decreased 1000–1400 ppm in drinking water or 19–39 mg/kg intra-
testosterone and LH levels, decreased testicular weight, peritoneally, pregnancy rate decreased with increased
and increase in the production of abnormal immobile frequency of atretic follicles; after pregnancy, failure
spermatozoa, whereas the parental F0 group did not of pregnancy increased with increased rate of uterine
present with such reproductive abnormalities (Muselin resorption and decrease in the number of viable fetuses
et al., 2016). In rats that were injected with Al chloride and implantation sites (Mohammed et al., 2008). Fu et al.
(4.125 pmole) in artificial cerebrospinal fluid via the lat- (2014) reported that Al exposure damaged ovarian struc-
eral ventricle, there were significant decreases in serum ture, disrupted metabolism of iron, zinc and copper in the
FSH, LH and testosterone levels, and reduction in sperm ovary and decreased the activities of ovarian ATPases and
count from the vas deferens and epididymides (Shahraki expressions of androgenic receptors for FSH and LH; and
et al., 2008). Bank voles (Myodes glareolus) exposed to could consequently lead to infertility due to inhibition of
Al produced lower quality and quantity of sperm than ovulation and development of corpus luteum. Exposure
normal, but reproductive capacity was not significantly to Al during mouse pregnancy resulted in reduced fetal
affected in females (Miska-Schramm et al., 2017). After weight and increased frequency of external anomalies in
intraperitoneal treatment at 50 mg/kg for 20 days, blood fetuses (Malekshah et al., 2005) and fetal micronucleated
testosterone and LH levels were increased in male rats, erythrocytes (D’Souza et al., 2014). Khalaf et al. (2007)
but FSH level was not affected (Resa and Palan, 2006). reported perinatal and postnatal adverse effects of Al
Khattab et al. (2010) reported that administration of Al exposure on fetuses and neonates during gestation and
chloride (20 mg/kg) to male rats via gavage for 70 days lactation of female rats. The hepatic toxicity of Al chloride
caused fertility disturbances and testicular dysfunction. was also reported in pregnant rats and their offspring
Other reports showed that Al induced decrease in sperm with observation of decreased fetal weight and size
counts, motility and viability, with increase in dead and (Mestaghanmi et al., 2003). Exposed embryonic chicks
abnormal sperm counts (Bataineh et al., 1998; Guo et and rat fetuses developed congenital myocardial defects
al., 2005; Yousef et al., 2007; Yousef and Salama, 2009; (Wang et al., 2012; El Mazoudy and Bekhet, 2016).

Figure 8. Reproductive and developmental disorders in aluminium toxicosis.

ISSN: 1337-6853 (print version) | 1337-9569 (electronic version)


Interdisciplinary Toxicology. 2019; Vol. 12(2): 45–70 59
Full-text also available online on PubMed Central

On the whole, Al toxicosis caused lesions in the testes


and ovaries resulting in impairment of spermatogenesis
and ovarian function related to ovulation, with the con-
sequence of reproductive inefficiency associated with
pregnancy failures and poor fetal development (Figure 8).

Hepato-renal and pancreatic effects


Al causes oxidative injuries to the kidney and liver leading
to tissue degeneration and necrosis, and associated serum
biochemical derangements (Nikolov et al., 2010; Mailloux
et al., 2011; Bai et al., 2012; Li et al., 2015; Xu et al., 2017).
Abdel-Wahab (2012) reported a significant increase
in the activities of alanine aminotransferase (ALT),
alkaline phosphatase (ALP), aspartate aminotransferase
(AST) and total bilirubin, as well as increased serum
urea and creatinine levels after oral administration of
20 mg/kg of Al chloride for 30 days in experimental rats.
Ingestion of aluminium phosphide pellets was reported Figure 9. Photomicrograph of pancreas of aluminium chloride-
treated rat showing coagulative necrosis of the pancreatic islet
to induce acute pancreatitis in one patient (Verma et al., tissue with disorganization of its architecture (H & E, x 400) From
2007). Rats had moderate pancreatic islet necrosis after Igwenagu et al. (2019).
intermediate oral exposure (50 mg/kg for 28 days) to Al
chloride (Figure 9) which was associated with impaired
fasting blood glucose and impaired oral glucose toler-
ance (Igwenagu, 2017; Igwenagu et al., 2019). Rats treated the biological characteristics of breast epithelial cells
intra-peritoneally with Al chloride at 10 mg/kg for 30 and carcinogenesis is considered a probable outcome
days had significantly increased fasting blood glucose, (Pineau et al., 2014). The content of Al in breast tissues
serum insulin level and insulin resistance index on days from mastectomies are being efficiently and accurately
10 and 20 of treatment, but as treatment progressed to estimated in order to properly assess the involvement of
day 30, serum insulin level had decreased, indicating that Al in the aetiology of breast cancer (House et al., 2013).
pancreatic β-cell function decreased as pancreatic dam- Current evidence suggests that Al can induce DNA dam-
age occurred with progression of treatment (Wei et al., age in human breast epithelial cells and subsequently
2018). The hepatic and pancreatic lesions cause changes induce proliferation of the cells (Darbre et al., 2013a, b).
in metabolism (Figure 5) which result in hyperglycaemia, Thus, Al may increase the risk of breast cancer by act-
hypoproteinaemia, hyperlipidaemia, hypercholester- ing as a metalloestogen (Darbre, 2016). The migratory
olaemia and hypertriglyceridaemia (Omar et al., 2003; and invasive properties of oestogen-responsive MCF-7
Kowalczyk et al., 2004; Türkez et al., 2011; Abdel-Wahab, human breast cancer cells were increased in the pres-
2012; Belaïd-Nouira et al., 2013). ence of Al (Darbre et al., 2013a). Long-term Al exposure
also increased the migration of oestrogen-unresponsive
Mammary gland or breast effects MDA-MB-231 human breast cancer cells in culture where
Breast cancers and cysts are mammary gland conditions their expression of matrix metalloproteinases (MMP9/14)
where emerging evidence are suggesting that Al may be was increased (Bakir and Darbre, 2015).
involved in their causation (Darbre, 2016). Al chlorohy-
drate in antiperspirant cosmetics and other underarm
cosmetic products may be an important source of Al Diagnosis and treatment of
exposure (Pineau et al., 2014; Linhart et al., 2017). In a aluminium intoxication
case control study (Linhart et al., 2017), the use of under-
arm cosmetic products containing Al was significantly Al can be measured in the blood, bone, urine, and feces to
associated with breast cancer incidence and the Al levels confirm Al load and association with toxicosis. A variety
in breast tissues were significantly higher in breast cancer of analytical methods have been used to measure Al
cases than controls (5.8 versus 3.8 nmol/g). Breast cancer levels in biological materials and they include accelerator
patients had higher levels of Al in breast tissues than in mass spectroscopy, graphite furnace atomic absorption
blood serum (Darbre et al., 2013b). There were higher spectrometry, flame atomic absorption spectrometry,
levels of Al in nipple aspirates of cancer patients than electro-thermal atomic absorption spectrometry, neutron
healthy controls and higher Al levels in breast cyst fluid activation analysis, inductively coupled plasma atomic
than serum or milk (Darbre et al., 2011). The Al contents emission spectrometry, inductively coupled plasma mass
of nipple aspirates of breast cancer patients correlated spectrometry, and laser microprobe mass spectrometry
with biomarkers of oxidative stress and inflammation (Maitani et al., 1994; Owen et al., 1994; Van Landeghem
in the breast microenvironment (Mannello et al., 2013). et al., 1994; Razniewska and Trzcinka-Ochocka, 2003).
The Al accumulating in the breast tissue may influence Contamination is a major problem encountered in the

Copyright © 2019 SETOX & Institute of Experimental Pharmacology and Toxicology, CEM SASc.
60 Aluminium toxicosis
Ikechukwu Onyebuchi Igbokwe, Ephraim Igwenagu, Nanacha Afifi Igbokwe

analysis of Al by all methods except that using radioactive intoxication (Omar et al., 2003; Abubakar et al., 2004;
26Al. When using the other methods, all items used dur- Fyiad, 2007; Turkez et al., 2011). Other researchers have
ing collection, preparation, and assay should be checked used plant extracts of fenugreek seed, grape seed, ginger,
for Al contribution to the procedure. wheat grass powder, black tea, Allium cepa, Caesalpinia
Treatment of Al intoxication is done with the chelat- crista, Arthrophytum (Hammada scoparia), Moringa
ing agent, deferoxamine, which is a colourless crystalline oleifera and Celastrus paniculatus to ameliorate the toxi-
base, produced by the bacterium, Streptomyces pilosus. cosis caused by Al exposure (Khattab et al., 2010; Hasseeb
Structurally, it is composed of one molecule of acetic et al., 2011; Osman et al., 2012; Belaïd-Nouira et al., 2013;
acid, two molecules of succinic acid and three molecules Sumathi et al., 2013; Bitra et al., 2014; Osama et al., 2014;
of 1-amino-5 hydroxylamine pentane (Keberle, 1964). Mathiyazahan et al., 2015; Singh and Goel, 2015; Taїr et
Deferoxamine is mainly used as an iron-chelating agent al., 2016; Ravi et al., 2018). The neuronal death in the hip-
to treat iron overload. But due to the chemical similar- pocampus of the brain associated with neurodegeneration
ity between Al and iron, it can also successfully mop-up in rats caused by Al exposure was attenuated by quercetin
excess Al from the body (Day, 1986; Martin et al., 1987). (Sharma et al., 2016). Ginsenoside Rb1 was reported
Deferoxamine administered intravenously has been to prevent Al-induced oxidative stress and reverse the
shown to reduce the body Al load and to ameliorate injury osteoblast viability and growth after impairment by Al
to the bone and brain in patients receiving hemodialysis (Zhu et al., 2016a). Chlorogenic acid was effective as a
and peritoneal dialysis (Malluche et al., 1984). It has also chelating agent and antioxidant in protection against the
been used successfully to treat Al toxicity in children toxicity of Al (Wang et al., 2018; Cheng et al., 2017, 2019).
(Warady et al., 1986; Ogborn et al., 1991). Deferoxamine Chenodeoxycholic acid ameliorated the neurotoxic effect
therapy seems beneficial for those with established Al of Al by improving insulin sensitivity (Bazzari et al., 2019).
toxicity; however, this therapy is not without hazards. Türkez et al. (2010) reported that propolis prevented the
It may cause allergic reactions such as pruritus, wheals genetic and hepatic damages induced by Al intoxication.
and anaphylaxis. Other adverse effects include dysuria, On the whole, the approach to the treatment of Al toxi-
abdominal discomfort, diarrhea, fever, leg cramps, cata- cosis after diagnosis involves strategies that include the
ract, and tachycardia (Klaassen, 1990). following: prevention of Al intake, reduction of Al absorp-
Malic acid is also a potent chelator of Al used in treat- tion, increasing Al excretion, maintaining functional kid-
ment of Al intoxication (Domingo et al., 1988). Treatment neys, reducing Al load by chelation with chelating agents
with malic acid has been reported to greatly increase and amelioration of toxic effects with antioxidants and
the fecal and urinary excretion of Al and reduce the other agents that reduce toxicity (Figure 10)
concentration of Al present in various organs and tissues
(Rim, 2007; Crisponi et al., 2012; Al-Qayim et al., 2014).
Other chelating agents such as citric, malonic, oxalic, and General perspective and conclusion
succinic acids have been used experimentally to reduce
aluminum load in rats and mice (Domingo et al., 1988). This review has provided an overview of the pathologic
Antioxidants and free radical scavengers such as basis of Al toxicosis. The association of Al intoxications
selenium, melatonin, boric acid and vitamin C have been with various pathologic syndromes and pathogenic
employed experimentally to ameliorate the deleteri- mechanisms linked to toxic actions may provide avenues
ous effects of free radicals produced as a result of Al for strategic interventions. The study of these pathologies

Figure 10. Prevention and treatment of aluminium toxicosis.

ISSN: 1337-6853 (print version) | 1337-9569 (electronic version)


Interdisciplinary Toxicology. 2019; Vol. 12(2): 45–70 61
Full-text also available online on PubMed Central

have received recent attention in epidemiological surveys The systemic effects caused by Al toxicosis are diverse
in regard to some human diseases such as Alzheimer’s and multifaceted, but co-morbidities from multisystemic
disease, autism, osteoporosis, diabetes mellitus, inflam- toxicosis are rarely reported in epidemiological cohorts.
matory bowel disease and others mentioned in the The convergence of toxic actions to engage multiple organ
review. We have reviewed the process of Al intoxication, systems in an individual is often an observation in experi-
toxic actions and systemic effects with an exploratory mental animal models and lacks validity in observational
approach to provide the subsequent highlights of the studies in human or animal populations. The possible
review in this section. action of Al in the pathogenesis of diabetes mellitus and
After the intake of Al and its deposition in tissues, the concurrence of neurological disorders associated with
the cell is the primary target of the toxic action of Al, Alzheimer’s disease and other dementias (Arnold et al.,
where the ion interacts with the plasma membrane 2018) point to the common cellular basis of the patho-
moieties, cytoplasmic biomolecules, mitochondria genesis of both metabolic and cognitive disorders, which
and nuclear structures. The major toxic action of Al is can arise from toxic actions of Al. Longitudinal studies in
to generate oxidative stress by producing reactive free the future may reveal co-morbidities and multisystemic
radicals which can overwhelm the antioxidant defenses toxicosis in Al-loaded individuals in locations where there
of the cell to perpetrate cellular injuries. The oxidative is high risk of Al exposure.
injuries emanate from the oxidation of proteins, lipids Al-induced oxidative stress with the metabolic defects
and nucleotides, which result in generation of altered that accompanies it may incidentally be the crux of the
functional biomolecules with defective operational toxicosis, to the extent that the use of antioxidant agents
capabilities towards cellular homeostasis. The disrup- forms the fundamental basis for therapeutic interven-
tion of homeostatic environments of the cell has the tions apart from chelating drugs. Chenodeoxycholic acid
capacity to change the semi-permeability and receptor improved insulin sensitivity to ameliorate the neurotoxic
functions of the plasma membrane, alter the reactivity of effect of Al (Bazzari et al., 2019). As new researches on
metabolic intermediary molecules and the functions of the cellular mechanisms in the toxicosis continue to be
enzymes and cofactors, and breach the energetic profile further elucidated through in vitro and in vivo studies of
and synthetic infrastructure at transcriptional and post- the metallic toxicant, new therapies against the toxicosis
transcriptional stages. In Al toxicosis, we identified the should also focus on alleviating the known aberrations
inhibition of cellular viability and function of neurons, in signaling pathways, synthetic and secretory functions,
osteoblasts, endocrine cells, lymphocytes, macrophages, cellular energetics and membrane integrity.
erythrocytes, erythroid cells, enterocytes, myocytes,
germinal cells, and pulmonary alveolar, hepatic, renal
and pancreatic islet cells. Progenitor or blast cells were
not able to proliferate, differentiate and function in REFERENCES
accordance with their genetic resources. The secretory
functions of specialized cells were impaired and several Abd-Elhady RM, Elsheikh AM, Khalifa AE. (2013). Anti-amnestic properties of
signaling pathways were recruited in abnormal chemico- Ginkgo biloba extract on impaired memory function induced by aluminum
in rats. Int J Dev Neurosci 31(7): 398–607.
biological settings. Cytokine expression was accelerated
Abdel-Wahab WM. (2012). AlCl3-induced toxicity and oxidative stress in liver
by oxidative cellular injuries to initiate inflammatory of male rats: protection by melatonin. Life Sci J 9: 1173–1182.
processes and alter immune responses that support Abder-Rahman H. (1999). Effect of aluminum phosphide on blood glucose
inflammation and immunosuppression, respectively. level. Vet Hum Toxicol 41: 31–32.
There were Al-induced endocrine disruptions and altered Abedini M, Fatehi F, Tabrizi N. (2014). Ischemic stroke as a rare manifestation
sensitivities to hormones such as insulin, parathyroid of aluminium phosphide poisoning: a case report. Acta Med Iran 52(12):
947–949.
hormone and hormonal vitamin D.
Abraham TM, Fox CS. (2013). Implications of rising prediabetes prevalence.
In Al toxicosis, the cellular structures were damaged Diabetes Care 36(8): 2139–2141.
by molecular mechanisms which cause degenerative Abubakar MG, Taylor A, Ferns GA. (2004). The effects of aluminium and sele-
changes (from lipid and amyloid depositions), cell death nium supplementation on brain and liver antioxidant status in the rat. Afr
by apoptosis or necrosis, and dysplasia from genetically J Biotechnol 3: 88–93.
driven cell growth abnormalities. Cellular degeneration Adzersen KH, Becker N, Steindorf K,Frentzel-Beyme R. (2003). Cancer mor-
tality in a cohort of male German iron foundry workers. Am J Ind Med 43:
occurred in nervous tissue, liver and kidney. Apoptosis 295–305.
was associated with damage to immune cells, erythroid Afridi HI, Kazi TG, Kazi N, Baig JA, Jamali MK, Arain MB, Sarfraz RA, Sheikh HU,
cells, erythrocytes, osteoblasts and germinal cells. Kandhro GA, Shah AQ. (2009). Status of essential trace metals in biological
Necrosis was encountered in pancreatic islet, liver, kid- samples of diabetic mother and their neonates. Arch Gynecol Obstet 280:
415–423.
ney, neurons and muscles. Dysplasia from chromosomal
Afridi HI, Talpur FN, Kazi TG, Brabazon D. (2015). Effect of trace and toxic ele-
aberrations was associated with developmental defects, ments of different brands of cigarettes on the essential elemental status of
teratogenesis and growth abnormalities in fetuses and Irish referent and diabetic mellitus consumers. Trace Elem Res 167(2): 209–
mammary epithelial cells. Mutagenesis, cell prolif- 224.
eration and impaired mitosis in Al toxicosis are gray Aguilar MV, Saavedra P, Arrieta FJ, Mateos CJ, Gonzalez MJ, Meseguer I, Mar-
tinez-Para MC. (2007). Plasma mineral content in type-2 diabetic patients
areas requiring clarification because of the observed and their association with the metabolic syndrome. Ann Nutr Metab 51:
antithesis. 402–406.

Copyright © 2019 SETOX & Institute of Experimental Pharmacology and Toxicology, CEM SASc.
62 Aluminium toxicosis
Ikechukwu Onyebuchi Igbokwe, Ephraim Igwenagu, Nanacha Afifi Igbokwe

Ahmad W. (2013). Overlapped metabolic and therapeutic links between Al- Belaïd-Nouira Y, Bakhta H, Haouas Z, Flehi-Slim I, Cheikh HB. (2013). Fenu-
zheimer’s disease and diabetes. Mol Neurobiol 47(1): 399–424. greek seeds reduce aluminum toxicity associated with renal failure in rats.
Ahn HW, Fulton B, Moxon D, Jeffery EH. (1995). Interactive effects of fluoride Nutr Res Pract 7: 466–474.
and aluminum uptake and accumulation in bones of rabbits administered Biego GH, Joyeux M, Hartemann P, Debry G. (1998). Daily intake of essential
both agents in their drinking water. J Toxicol Environ Health 44: 337–350. minerals and metallic micropollutants from foods in France. Sci Total Envi-
ron 217: 27–36.
Akhlaghi F, Bagheri SM, Rajabi O. (2012). A comparative study of relation-
ship between micronutrients and gestational diabetes. ISRN Obstet Gynecol Bilkei-Gorzó A. (1993). Neurotoxic effect of enteral aluminum. Food Chem
2012, Article ID 470419, 4 pages, http: //dx.doi.org/10.5402/2012/470419. Toxicol 31: 357–361.
Akinola OB, Biliaminu SA, Adedeji OG, Oluwaseun BS, Olawoyin OM, Adelabu Bitra VR, Rapaka D, Mathala N, Akula A. (2014). Effect of wheat grass powder
TA. (2016). Combined effects of chronic hyperglycaemia and oral alumin- on aluminum induced Alzheimer’s disease in Wister rats. Asian Pac J Trop
ium intoxication on testicular tissue and some male reproductive paame- Med 7(S1): S278–S281.
ters in Wistar rats. Andrologia 48(7): 779–786. Blaylock RL. (2012). Aluminium induced immunoexcitotoxicity in
Al-Qayim MAJ, Ghali LS, Al-Azwai TS. (2014). Comparative effects of propolis neurodevelopmental and neurodegenerative disorders. Current Inorg
and malic acid on hematological parameters of aluminum exposed male Chem 2: 1–8.
rats. Global J Bio-Sci Biotechnol 3: 6–11. Bogdanović M, Janeva AB, Bulat P. (2008). Histopathological changes in rat
liver after a single high dose of aluminium. Arch Ind Hyg Toxicol 59: 97–101.
Alfrey AC. (1980). Aluminum metabolism in uremia. Neurotoxicology 1: 43–53.
Bondy SC. (2016). Low levels of aluminum can lead to behavioral and mor-
Alfrey AC. (1993). Aluminum toxicity in patients with chronic renal failure.
phological changes associated with Alzheimer’s disease and age-related
Ther Drug Monit 15(6): 593–597.
neurodegeneration. Neurotoxicology 52: 222–229.
Alfrey AC, Solomons C. (1976). Bone pyrophosphate in uremia and its associa-
Bondy SC, Truong A. (1999). Potentiation of beta-folding of β-amyloid pep-
tion with extraosseous calcification. J Clin Invest 57: 700–705. tide 25-35 by aluminum salts. Neurosci Lett 267(1): 25–28.
Alter P, Grimm W, Maisch B. (2001). Lethal heart failure caused by aluminium Boran AM, Al-Khatib AJ, Alanazi BS, Massadeh AM (2013). Investigation of alu-
phosphide poisoning. Intensive Care Med 27: 327–331. minum toxicity among workers in aluminum industry sector. Eur Scient J 9:
Anane R, Creppy EE. (2001). Lipid peroxidation as pathway of aluminium cy- 440–451.
totoxicity in human skin fibroblast cultures: prevention by superoxide Boyce BF, Mocan MZ, Junor BJR. (1986). Toxic effect of aluminium and other
dismutase+catalase and vitamins E and C. Hum Exp Toxicol 20: 477–481. substances on bone turnover. Nefrologia 6: 103–108.
Andreoli SP. (1990). Aluminum levels in children with chronic renal failure Brown S, Savory J, Wills MR. (1987). Absorption of aluminum from citrate or
who consume low phosphorus infant formula. J Pediatr 116: 282–285. chloride in everted rat gut sac. Clin Chem 33: 933–934.
Anon (1982). Drinking Water and Health. National Research Council, National Browne BA, McColl JG, Driscoll CT. (1990). Aluminum speciation using morin:
Academy Press, Washington, DC 4: 155–201. I. morin and its complexes with aluminum. J Environ Qual 19: 65–82.
Anon (2008a). Aluminum and compounds. Hazardous Substances Data Bank. Buraimoh AA, Ojo SA. (2012). Effects of aluminium chloride exposure on the
Bethesda, MD: National Library of Medicine. http: //toxnet.nlm.nih.gov/cgi- histology of the small intestine of wistar rats. Int J Appl Sci Technol 2(10):
bin/sis/htmlgen?HSDB Accessed 9 January 2016. 123–132.
Anon (2008b). Safety of aluminium from dietary intake. EFSA J 754: 1–34. Buraimoh AA, Ojo SA. (2013). Effects of aluminium chloride exposure on the
Anon (2008c). Toxicological profile for aluminum. Atlanta, GA, United States histology of lungs of wistar rats. J Appl Pharm Sci 3: 108–112.
Department of Health and Human Services, Public Health Service, Agency Burge PS, Scott JA, McCoach J. (2000). Occupational asthma caused by alumi-
for Toxic Substances and Disease Registry. http: //www.atsdr.cdc.gov/tox- num. Allergy 55(8): 77–780.
profiles/tp22.pdf Accessed 27 January 2016. Burgoin BP. (1992). Alumino-silicate content in calcium supplements derived
Anon (2011). On the evaluation of a new study to the bioavailability of alu- from various carbonate deposits. Bull Environ Contam Toxicol 48: 803–808.
minium in food. EFSA J 9: 1–16. Bushinsky DA, Sprague SM, Hallegot P, Girod C, Chabala JM, Levi-Setti R.
Arain MS, Afridi HI, Kazi TG, Talpur FN, Arain MB, Kazi A, Arain SA, Ali J. (2015). (1995). Effect of aluminum on bone surface ion composition. J Bone Miner
Correlation of aluminum and manganese concentrations in scalp hair sam- Res 10: 1988–1997.
ples of patients having neurological disorders. Environ Monit Assess 187(2): Cao Z, Fu Y, Sun X, Zhang Q, Xu F, Li Y. (2016). Aluminum inhibits osteoblastic
10. differentiation through inactivation of Wnt/β-catenin signaling pathway in
Arnold SE, Arvanitakis Z, Macauley-Rambach SL, Koenig AM, Wang H, Ahima rat osteoblasts. Environ Toxicol Pharmacol 42: 198–204.
RS, Craft S, Gandy S, Buettner C, Stoeckel LE, Holtzman DM, Nathan DM. Campbell A, Bondy SC. (2000). Aluminum induced oxidative events and its
(2018). Brain insulin resistance in type 2 diabetes and Alzheimer’s disease: relation to inflammation: a role for the metal in Alzheimer’s disease. Cell
concept and conumdrums. Nature Rev Neurol 14(3): 168–181. Mol Biol 46(4): 721–730.
Bacon R, Tavill A, Brittenham G, Park C, Recknagel R. (1983). Hepatic lipid per- Campbell A, Prasad KN, Bondy SC. (1999). Aluminum-induced oxidative
oxidation in vivo in rats with chronic iron overload. J Clin Invest 71: 429– events in cell lines: glioma are more responsive than neuroblastoma. Free
439. Rad Biol Med 26: 1166–1671.
Bai C, Wang F, Zhao H, Li Y. (2012). Effects of subchronic aluminum exposure Campbell PGC, Hansen HJ, Dubreuil B, Nelson WO. (1992). Geochemistry of
on liver function in rats. J Northeast Agric Univ 19(2): 62–65. Quebec north shore salmon rivers during snowmelt: Organic acid pulse
and aluminum mobilization. Can J Fish Aquat Sci 49: 1938–1952.
Bakir A, Darbre PD. (2015). Effect of aluminium on migration of oestrogen un-
responsive MDA-MB-231 human breast cancer cells in culture. J Inorg Bio- Cannata JB, Briggs JD, Junor BJ, Fell GS, Beastall G. (1983). Effect of acute alu-
chem 152: 180–185. minium overload on calcium and parathyroid-hormone metabolism. Lan-
cet 1(8323): 501–503.
Bank WA, Kastin AJ, Maness LM, Huang W, Jaspan JB. (1995). Permeability of
the blood-brain barrier by amylin. Life Sci 57(22): 1993–2001. Capdevielle MC, Scanes CG. (1995). Effect of dietary acid or aluminum on
growth and growth-related hormones in mallard ducklings (Anas platy-
Bataineh H, Al-Hamood MH, Elbetieha AM. (1998). Assessment of aggressive, rhynchos). Arch Environ Contam Toxicol 29(4): 462–468.
sexual behavior and fertility in adult rat following long term ingestion of
Cech I, Montera J. (2000). Spatial variations in total aluminum concentrations
four industrial metals salts. Hum Exp Toxicol 17: 570–579.
in drinking water supplies studied by geographic information system (GIS)
Bazzari F, Abdallah DM, El-Abhar HS. (2019). Chenodeoxycholic acid amelio- methods. Water Res 34: 2703–2712.
rates AlCl3-induced Alzheimer’s disease neurotoxicity and cognitive dete-
Chafe R, Aslanov R, Sarkar A, Gregory P, Comeau A, Newhook LA. (2018). As-
rioration via enhanced insulin signaling in rats. Molecules 24(10): 1992.
sociation of type 1 diabetes and concentrations of drinking water compo-
Bazzoni GB, Bollini AN, Hernández GN, Contini MC, Rasia ML (2005). In vivo nents in Newfoundland and Labrador, Canada. BMJ Open Diabetes Res Care
effect of aluminium upon the physical properties of the erythrocyte mem- 6(1): e000466.
brane. J Inorg Biochem 99(3): 822–827. Chappard D, Bizot P, Mabilleau G, Hubert L. (2016). Aluminum and bone: re-
Becaria A, Campbell A, Bondy SC. (2002). Aluminum as a toxicant. Toxicol Ind view of new clinical circumstances associated with Al(3+) deposition in the
Health 10(7): 309–320. calcified matrix of bone. Morphologie 100(329): 95–105.

ISSN: 1337-6853 (print version) | 1337-9569 (electronic version)


Interdisciplinary Toxicology. 2019; Vol. 12(2): 45–70 63
Full-text also available online on PubMed Central

Chen BB, Zeng Y, Hu B. (2010). Study on speciation of aluminum in human Day JP. (1986). Chemical aspects of aluminum chelation by desferrioxamine,
serum using zwitterionic bile acid derivative dynamically coated C18 col- in Aluminum and other trace elements in renal disease (Taylor A ed) pp. 184–
umn HPLC separation with UV and on-line ICP-MS detection. Talanta 81: 192, Baillere-Tindall, London.
180–186. Day JP, Barker J, Evans LJA, Perks J, Seabright PJ, Ackrill P, Lilley JS, Drumm PV,
Chen WJ, Monnat RJ Jr, Chen M, Mottet NK. (1978). Aluminum induced pul- Newton GWA. (1991). Aluminum absorption studied by 26Al tracer. Lancet
monary granulomatosis. Hum Pathol 9(6): 705–711. 337: 1345.
Chen YW, Yang CY, Huang CF, Hung DZ, Leung YM, Liu SH. (2009). Heavy met- de Chambrun GP, Body-Malapel M, Frey-Wagner I, Djouina M, Deknuydt F,
als, islet function and diabetes development. Islets 1: 169–176. Atrott K, Esquerre N, Altare F, Neut C, Arrieta MC, Kanneganti T-D, Rogler G,
Chen Z, Zhong C. (2014). Oxidative stress in Alzheimer’s disease. Neurosci Bull Colombel JF, Cortot A, Desreumaux P, Vignal C. (2014). Aluminum enhances
30(2): 271–281. inflammation and decreases mucosal healing in experimental colitis in
mice. Mucosal Immunol 7(3): 589–601.
Cheng D, Tang J, Wang X, Zhang X, Wang S. (2018). Effect of aluminum (Al)
speciation on erythrocytic antioxidant defense process: Correlations be- de la Monte SM, Wand JR. (2008). Alzheimer’s disease is type 3 diabetes- Evi-
tween lipid membrane peroxidation and morphological characteristics. dence reviewed. J Diabetes Sci Technol 2(6): 1101–1113.
Ecotoxicol Environ Saf 157: 201–206. Delhaize E, Ryan RP. (1995). Aluminum toxicity and tolerance in plants. Plant
Cheng D, Zhang X, Tang J, Kong Y, Wang X, Wang S. (2019). Chlorogenic acid Physiol 107: 315–321.
protects against aluminum toxicity via MAPK/Akt signaling pathway in mu- Deugnier JL. (2003). Iron and liver cancer. Alcohol 30: 145–150.
rine RAW264.7 macrophages. J Inorg Biochem 190: 113–120. DeVoto E, Yokel RA. (1994). The biological speciation and toxicokinetics of
Cheng D, Zhang X, Xu L, Li X, Hou L, Wang C. (2017). Protective and prophy- aluminum. Environ Health Perspect 102: 940–951.
lactic effects of chlorogenic acid on aluminum-induced acute hepatotoxic- de Vuyst P, Dumortier P, Schandené L, Estenne M, Verhest A, Yernault JC.
ity in mice. Chem Biol Interact 273: 125–132. (1987). Sarcoid-like lung granulomatosis induced by aluminum dusts. Am
Chines A, Pacifici R. (1990). Antacid and sucralfate-induced hypophospha- Rev Respir Dis 135(2): 493–497.
temic osteomalacia: A case report and review of the literature. Calcif Tissue Díaz-Corte C, Fernández-Martín JL, Barreto S, Gómez C, Fernández-Coto T,
Int 47: 291–295. Braga S, Cannata JB. (2001). Effect of aluminium load on parathyroid hor-
Chinoy NJ, Patel TN. (2001). Effect of sodium fluoride and aluminium chloride mone synthesis. Nephrol Dial Transplant 16(4): 742–745.
on ovary and uterus of mice and their reversal by some antidotes. Fluoride
Dixon RL, Sherins RJ, Lee IP. (1979). Assessment of environmental factors af-
34(1): 9–20.
fecting male fertility. Environ Health Perspect 30: 53–68.
Chmielnicka J, Nasiadek M, Pinkowski R, Paradowski M. (1994). Disturbances
Dokken BB, Saengsirisuwan V, Kim JS, Teachey MK, Henriksen EJ. (2008). Oxi-
of morphological parameters in blood of rats orally exposed to aluminum
dative stress-induced insulin resistance in rat skeletal muscle: role of gly-
chloride. Biol Trace Elem Res 42: 191–199.
cogen synthase kinase-3. American Jounal of Physiology. Endocrinol Metab
Chopra JS, Kalra OP, Malik VS, Sharma R, Chandna A. (1986). Aluminum phos- 294(3): E615–621.
phide poisoning: A prospective study of 16 cases in one year. Postgrad Med
Domingo JL. (1995). Reproductive and developmental toxicity of aluminum:
J 62: 1113–1115.
a review. Neurotoxicology 17(4): 515–521.
Christie H, MacKay RJ, Fisher AM. (1963). Pulmonary effects of inhalation of
Domingo JL, Gomez M, Liobet JM, Corbella J. (1988). Comparative effects of
aluminum by rats and hamsters. AIHA J (Fairfax, Va) 24: 47–56.
several chelating agents on the toxicity, distribution, and excretion of alu-
Codling EE, Chaney RL,Mulchi CL. (2002). Biomass yield and phosphorus minum. Hum Toxicol 7: 259–262.
availability to wheat grown on high phosphorus soils amended with phos-
Domingo JL, Llobet JM, Gomez M, Tomas JM, Corbella J. (1987). Nutritional
phate inactivating residues. I. Drinking water treatment residue. Commun
and toxicological effects of short-term ingestion of aluminum by the rat.
Soil Sci Plant Anal 33: 1039–1061.
Res Commun Chem Pathol Pharmacol 56: 409–419.
Colomina MT, Peris-Sampedro F. (2017). Aluminum and Alzheimer’s disease.
D’Souza SP, Vijayalaxmi KK, Prashantha N. (2014). Assessment of genotoxic-
Adv Neurobiol 18: 183–197.
ity of aluminium acetate in bone marrow, male germ cells and fetal cells
Colomina MT, Roig JL, Sanchez DJ, Domingo JL. (2002). Influence of age on of Swiss albino mice. Mutat Res Genet Toxicol Environ Mutagen 766: 16–22.
aluminum-induced neurobehavioral effects and morphological changes in
Drüeke T, Touam M, Lacour B. (1986). Aluminum-induced microcytic anemia
rat brain. Neurotoxicology 23: 775–781.
in experimental chronic renal failure. Nefrologia 6: 67–69.
Contini MC, Ferri A, Bernal CA, Carnovale CE. (2007). Study of iron homeosta-
Dunn MA, Ishizakie AS, Liew MYB, Too SL, Johnson N. (1995). Dietary alumi-
sis following partial hepatectomy in rats with chronic aluminum intoxica-
num chloride inhibits the ability of vitamin D to regulate intestinal calbin-
tion. Biol Trace Elem Res 115: 31–45.
din-D28k levels in chicks. J Trace Elem Exp Med 8: 47–57.
Cournot-Witmer G, Plachot JJ. (1990). Parathyroid glands in chronic alumi-
num intoxication. Ultrastruct Pathol 14(3): 211–219. Edwardson JA, Moore PB, Ferrier IN, Lilley JS, Newton GWA, Barker J, Templar
J, Day JP. (1993). Effect of silicon on gastrointestinal absorption of alumin-
Cox KA, Dunn MA. (2001). Aluminum toxicity alters the regulation of calbi- ium. Lancet 342: 211–212.
ndin-D28k protein and mRNA expression in chick intestine. J Nutr 131(7):
2007–2013. Eisenreich SJ. (1980). Atmospheric input of trace metals to Lake Michigan
(USA). Water Air Soil Pollut 13: 287–301.
Crisponi G, Nurchi VM, Bertolasi V, Remelli M, Faa G. (2012). Chelating agents
for human diseases related to aluminium overload. Coord Chem Rev 256: El Mazoudy RH, Bekhet GA. (2016). In ovo toxico-teratological effects of alu-
89–104. minum on embryonic chick heart and vascularization. Environ Sci Pollut Res
Int 23(21): 21947–21956.
Cronan CS, Schofield CL. (1979). Aluminum leeching response to acid precipi-
tation: Effect on high-elevation watersheds in the northeast. Science 204: Exley C. (2004). The proxidant activity of aluminium. Free Rad Biol Med 36(3):
304–306. 380–387.
Darbre PD. (2016). Aluminium and the human breast. Morphologie 100(329): Exley C. (2006). Aluminium and iron, but neither copper nor zinc, are key to
65–74. the precipitation of beta-sheets of A beta (42) in senile plaque cores in Al-
zheimer’s disease. J Alzheimers Dis 10: 173–177.
Darbre PD, Bakir A, Iskakova E. (2013a). Effect of aluminium on migratory and
invasive properties of MCF-7 human breast cancer cells in culture. J Inorg Exley C. (2013). Human exposure to aluminium. Environ Sci Process Impacts
Biochem 128: 245–249. 15: 1807–1816.
Darbre PD, Mannello F, Exley C. (2013b). Aluminium and breast cancer: Exley C. (2016). The toxicity of aluminium in humans. Morphologie 100(329):
sources of exposure, tissue measurmenets and mechanisms of toxicologi- 51–55.
cal actions of breast of breast biology. J Inorg Biochem 128: 257–261. Exley C, Begum A, Woolley MP, Bloor RN. (2006). Aluminum in tobacco and
Darbre PD, Pugazhendhi D, Mannello F. (2011). Aluminium and Human breast cannabis and smoking-related disease. Am J Med 119(3): 276.e9–11.
diseases. J Inorg Biochem 105(11): 1484–1488. Exley C, Birchall JD. (1992). The cellular toxicity of aliminium. J Theor Biol
Dasappa H, Fathima FN, Prabhkar R, Sarin S. (2015). Prevalence of diabetes 159(1): 83–98.
and prediabetes and assessment of risk factors in urban slums of Banga- Exley C, Birchall JD, Price N. (1994). Luminum inhibition of hexokinase activ-
lore. J Family Med Prim Care 4(3): 399–404. ity in vitro: A study in biological availability. J Inorg Biochem 54(4): 297–304.

Copyright © 2019 SETOX & Institute of Experimental Pharmacology and Toxicology, CEM SASc.
64 Aluminium toxicosis
Ikechukwu Onyebuchi Igbokwe, Ephraim Igwenagu, Nanacha Afifi Igbokwe

Exley C, House E. (2011). Aluminium in the human brain. Monatshefte fur Che- Golub MS, Han B, Keen CL, Gershwin ME. (1992). Effects of dietary aluminum
mie 142: 357–363. excess and manganese deficiency on neurobehavioral endpoints in adult
Exley C, Price NC, Kelly SM, Birchall JD. (1993). An interaction of β-amyloid mice. Toxicol Appl Pharmacol 112: 154–160.
with aluminium in vitro. FEBS Lett 324(3): 293–295. Gomes LS, Costa JR, Campos MS, Marques MR, Biancardi MF, Taboga SR,
Exley C, Siesjo P, Eriksson H. (2010). The immunobiology of aluminium adju- Ghedini PC, Santos FCA. (2019). Aluminum disrupts the prenatal develop-
vants: how do they really work? Trends Immunol 31: 103–109. ment of male and female gerbil prostrate (Meriones unguiculatus). Exp Mol
Pathol 107: 32–42.
Exley C, Swarbrick L, Gherardi RK, Authier FJ. (2009). A role for the body bur-
den of aluminium in vaccine-associated macrophagic myofasciitis and Gomez M, Domingo JL, Llobet JM, Tomas JM, Carbella J. (1986). Short-term
chronic fatigue syndrome. Med Hypotheses 72(2): 135–139. oral toxicity study of aluminum in rats. Arch Pharmacol Toxicol 12: 145–151.
Fanti P, Kindy MS, Mohapatra S, Klein J, Columbo G, Malluche HH. (1992). Gonzalez MA, Alvarez ML, Pisani GB. (2007). Involvement of oxidative stress
Dose-dependent effects of aluminum on osteocalcin synthesis in osteo- in the impairment in biliary secretory function induced by intraperitoneal
blast like ROS 17/2 cells in culture. Am J Physiol 263: E1113–E1118. administration of aluminum to rats. Biol Trace Elem Res 116: 329–348.
Farina M, Rotta LN, Soares FA, Jardim F, Jacques R, Souza DO, Rocha JB. Gonzelez-Suerez I, ÁLvarez-Hernendez D, Carrillo-Lepez N, Naves-Deaz M,
(2005). Hematological changes in rats chronically exposed to oral alumi- Fernendez-Marten JL, Cannata-Andea JB. (2005). Aluminum posttranscrip-
num. Toxicology 209: 29–37. tional regulation of parathyroid hormone synthesis: A role for the calcium-
sensing receptor. Kidney Int 68(6): 2484–2496.
Fernández MS. (2014). Human IAPP amyloidogenic properties and pancreatic
β-cell death. Cell Calcium 56(5): 416–427. Gorsky JE, Dietz AA, Spencer H. (1979). Metabolic balance of aluminum in
persons receiving aluminum antacids. Clin Chem 25: 244–248.
Feinroth M, Feinroth MV, Berlyne GM. (1984). Aluminum absorption in the rat
everted gut sac. Miner Electrolyte Metab 8: 29–35. Green BN, Johnson CD, Adams A. (2006). Writing narrative literature reviews
for peer-reviewed journals: secrets of the trade. J Chiropr Med 5(3): 101–117.
Feng W, Cui X, Liu B, Liu C, Xiao Y, Lu W, Guo H, He M, Zhang X, Yuan J, Chen
W,Wu T. (2015). Association of urinary metal profiles with altered glucose Greger JL. (1992). Dietary and other sources of aluminum intake. In: Alumi-
levels and diabetes risk: a population-based study in China. PLoS One 10: num in Biology and Medicine. Ciba Found Symp 169: 26–49.
1–11. Greger JL, Sutherland J. (1997). Aluminum exposure and metabolism. Critical
Filipek LH, Nordstrom DK, Ficklin WH. (1987). Interaction of acid mine drain- Review. Clin Lab Sci 34: 439–474.
age with waters and sediments of West Squaw Creek in the West Shasta Guo C, Lu Y, Hsu GSW. (2005). The influence of aluminum exposure on male
mining district, California. Environ Sci Technol 21: 388–396. reproduction and offspring in mice. Environ Toxicol Pharmacol 20: 135–141.
Flaten TP. (2001). Aluminium as a risk factor in Alzheimer’s disease, with em- Haglin L, Essén-Gustavsson B, Lindholm A. (1994). Hypophosphatemia in-
phasis on drinking water. Brain Res Bull 55(2): 187–196. duced by dietary aluminium hydroxide in growing pigs: effects on eryth-
Flaten TP. (2002). Aluminium in tea-concentrations, speciation and bioavail- rocytes, myocardium, skeletal muscle and liver. Acta Vet Scand 35(3): 263–
ability. Coord Chem Rev 228: 385–395. 271.
Florence AL, Gauthier A, Ponsar C, Van den Bosch de Aguilar P, Crichton RR. Han S, Lemire J, Appanna VP, Auger C, Castonguay Z, Appanna VD. (2013).
(1994). An experimental animal model of aluminum overload. Neurodegen- How aluminium, an intracellular ROS generator, promotes hepatic and
eration 3: 315–323. neurological diseases: the metabolic tale. Cell Biol Toxicol 29(2): 75–84.
Flores CR, Puga MP, Wrobel K, Sevilla MEG, Wrobel K. (2011). Trace elements Hangouche AJE, Fennich H, Alaika O, Dakka T, Raisouni Z, Oukerraj L,
status in diabetes mellitus type 2: Possible role of the interaction between Doghmi N, Cherti M. (2017). Reversible myocadial injury and intra-
molybdenum and copper in the progress of typical complications. Diabe- ventricular thrombus associated with aluminium phosphide poison-
tes Res Clin Pract 91: 333–341. ing. Case Rep Cardiol 2017, Article ID 6287015, 6 pages, https: //doi.
org/10.1155/2017/6287015.
Fogarty U, Perl D, Good P, Ensley S, Seawright A, Noonan J. (1998). A cluster
of equine granulomatous enteritis cases: the link with aluminium. Vet Hum Hansen AB, Ravnskjær L, Loft S, Andersen KK, Bräuner EV, Baastrup R, Yao C,
Toxicol 40(5): 297–305. Ketzel M, Becker T, Brandt J, Hertel O, Andersen ZJ. (2016). Long-term ex-
posure to fine particulate matter and incidence of diabetes in the Danish
Fu Y, Jia FB, Wang J, Song M, Liu SM, Li YF, Liu SZ, Bu QW. (2014). Effects of
sub-chronic aluminum chloride exposure on rat ovaries. Life Sci 100(1): 61– Nurse Cohort. Environ Int 91: 243–250.
66. Harris WR, Messori L. (2002). A comparative study of aluminum (III), gallium
Fyiad AA. (2007). Aluminium toxicity and oxidative damage reduction by (III), indium (III), and thallium (III) binding to human serum transferrin. Co-
melatonin in rats. J Appl Sci Res 3: 1210–1217. ord Chem Rev 228: 237–262.
Ganrot PO. (1986). Metabolism and possible health effects of aluminum. Envi- Hasan NA. (2009). Effects of trace elements on albumin and lipoprotein gly-
ron Health Perspect 65: 363–441. cation in diabetic retinopathy. Saudi Med J 30: 1263–1271.
Garbossa G, Galvez G, Castro ME, Nesse A. (1998). Oral aluminum adminis- Hasseeb MM, Al-Hizab AF, Hussein AY. (2011). A histopathologic study of the
tration to rats with normal renal function. 1. Impairment of erythropoiesis. protective effect of grape seed extract against experimental aluminum
Hum Exp Toxicol 17: 312–317. toxicosis in male rat. Scient J King Faisal Univ (Basic Appl Sci) 12: 283–297.
Garbossa G, Gutnisky A, Nesse A. (1996). Depressed erythroid progenitor cell Hayacibara MF, Queiroz CS, Tabchoury CPM,Cury JA. (2004). Fluoride and alu-
activity in aluminum overloaded mice. Miner Electrolyte Metab 22: 214–218. minum in teas and tea-based beverages. Rev Saude Publica 38: 100–105.
Garruto RM, Shankar SK, Yanagihara R, Salazar AM, Amyx HL, Gajdusek DC. He P, Zou Y, Hu Z. (2015). Advances in aluminum hydroxide-based adjuvant
(1989). Low-calcium, high-aluminum diet-induced motor neuron pathol- research and its mechanism. Hum Vaccin Immunother 11(2): 477–488.
ogy in cynomolgus monkeys. Acta Neuropathol 78: 210–219. Hem SL. (2002). Elimination of aluminum adjuvants. Vaccine 20: S40–43.
Gensemer RW, Playle RC. (1999). The bioavailability and toxicity of aluminum Hemadi M, Miquel G, Kahn PH, Chahine JME. (2003). Aluminum exchange be-
in aquatic environments. Crit Rev Environ Sci Technol 29(4): 315–450. tween citrate and human serum transferrin and interaction with transferrin
Gherardi RK, Aouizerate J, Cadusseau J, Yara S, Authier FJ. (2016). Aluminum receptor 1. Biochemistry 42: 3120–3130.
adjuvants of vaccines injected in muscle: Normal fate, pathology and asso- Henriksen EJ. (2010). Dysregulation of glycogen synthase kinase-3 in skeletal
ciated disease. Morphologie 100(329): 85–94. muscle and the etiology of insulin resistance and type 2 diabetes. Curr Dia-
Gherardi RK, Authier FJ. (2012). Macrophagic myofaciitis: characterization betes Rev 6(5): 285–293.
and pathophysiology. Lupus 21(2): 184–189. Henshaw PF, Bewtra JK, Biswas N. (1993). Occurrence of aluminum, lead, and
Glanz JM, Newcomer SR, Daley MF, McClure DL, Baxter RP, Jackson ML, Nale- trihalomethanes in drinking water from the Great Lakes. J Great Lakes Res
way AL, Lugg MM, DeStefano F. (2015). Cummulative and episodic vaccine 19: 521–532.
aluminum exposure in a population based cohort of young children. Vac- Herbert A, Sterling G, Abraham J, Corrin B. (1982). Desquamative interstitial
cine 33(48): 6736–6744. pneumonia in an aluminum welder. Hum Pathol 13(8): 694–699.
Golub MS, Germann SI. (2001). Long-term consequences of developmental Hernández G, Bollini A, Huarte M, Bazzoni G, Piehl L, Chiarotto M, Rubin de
exposure to aluminum in a suboptimal diet for growth and behavior of Celis E, Rasia M. (2008). In vitro effect of aluminium upon erythrocyte mem-
Swiss Webster mice. Neurotoxicol Teratol 23: 365–372. brane properties. Clin Hemorheol Microcirc 40(3): 191–205.

ISSN: 1337-6853 (print version) | 1337-9569 (electronic version)


Interdisciplinary Toxicology. 2019; Vol. 12(2): 45–70 65
Full-text also available online on PubMed Central

Hewitt CD, Savory J, Wills MR. (1990). Aspects of aluminium toxicity. Clin Lab Kamalov J, Carpenter DO, Birman I. (2011). Cytotoxicity of environmentally
Med 10: 403–422. relevant concentrations of aluminum in murine thymocytes and lympho-
His E, Beiras R, Seaman MN, Pagano G, Trieff NM. (1996). Sublethal and lethal cytes. J Toxicol 2011: 796719.
toxicity of aluminum industry effluents to early developmental stages of Kamanyire R, Murray V. (2003). Occupational exposures to fumigants. J Toxi-
the Crassostrea gigas oyster. Arch Environ Contam Toxicol 30: 335–339. col Clin Toxicol 41: 489–490.
Hofstetter JR, Vincent I, Bugiani O, Richter JA. (1987). Aluminum-induced de- Kandimalla R, Thirumala V, Reddy PH. (2017). Is Alzheimer’s disease a type 3
creases in choline acetyltransferase, tyrosine hydroxylase and glutamate diabetes? A critical appraisal. Biochim Biophys- Mol Basis Dis 1863(5): 1078–
decarboxylase in selected regions of rabbit brain. Neurochem Pathol 6(3): 1089.
177–193. Kandimalla R, Vallamkondu J, Corgiat EB, Gill KD. (2016). Understanding as-
Hogenesch H. (2013). Mechanism of immunopotentiation and safety of alu- pects of aluminum exposure in Alzheimer’s disease development. Brain
minum adjuvants. Frontiers Immunol 10: 406. Pathol 26(2): 139–154.
House E, Polwart A, Darbre P, Barr L, Metaxas G, Exley C. (2013). The alumin- Katz AC, Frank DW, Sauerhoff MW, Zwicker GM, Freudenthal RI. (1984). A
ium content of breast tissue taken from women with breast cancer. Med 6-month dietary toxicity study of acidic sodium aluminum phosphate in
Biol 27(4): 257–266. beagle dogs. Food Chem Toxicol 22: 7–9.
Huang W, Wang P, Shen T, Hu C, Han Y, Song M, Bian Y, Li Y, Zhu Y. (2017). Alu- Kawahara M, Kato-Negishi M. (2011). Link between aluminum and the patho-
minum trichloride inhibited osteoblastic proliferation and downregulated genesis of Alzheimer’s disease: The integration of the aluminum and amy-
the Wnt/β-catenin pathway. Biol Trace Elem Res 177(2): 323–330. loid cascade hypothesis. Int J Alzheimer’s Dis 2011: 276393.
Igbokwe NA. (2016). Characterization of the osmotic stability of Sahel goat Kawahara M, Konoha K, Nagata T, Sadakane Y. (2007). Aluminum and human
erythrocytes in ionic and non-ionic hypotonic media. PhD thesis, Depart- health: its intake, bioavailability and neurotoxicity. Biomed Res Trace Elem
ment of Physiology, Pharmacology and Biochemistry, Faculty of Veterinary 18: 211–220.
Medicine, University of Maiduguri, Maiduguri, Nigeria. Kazi TG, Afridi HI, Kazi N, Jamali MK, Arain MB, Jalbani N, Kandhro GA. (2008).
Igbokwe NA. (2018). A review of the factors that influence erythrocyte os- Copper, chromium, manganese, iron, nickel and zinc levels in biological
motic fragility. Sokoto J Vet Sci 16(4): 1–23. samples of diabetes mellitus patients. Biol Trace Elem Res 122: 1–18.
Igwenagu E. (2017). Pathologic effects of oral aluminium administration in Kazi TG, Jalbani N, Arain MB, Jamali MK, Afridi HI, Shah AQ. (2009). Determi-
non-diabetic and diabetic rats. MVSc dissertation, Department of Veteri- nation of toxic elements in different brands of cigarette by atomic absorp-
nary Pathology, University of Maiduguri, Maiduguri, Nigeria. tion spectrometry using ultrasonic assisted acid digestion. Environ Monit
Assess 154(1–4): 155–167.
Igwenagu E, Igbokwe IO, Egbe-Nwiyi TN. (2019). Fasting hyperglycaemia,
glucose intolerance and pancreatic islet necrosis in albino rats associated Keberle H. (1964). The biochemistry of DFO and its relation to iron metabo-
with subchronic oral aluminium chloride exposure. Comp Clin Pathol http: lism. Ann N Y Acad Sci 119: 758–776.
//dx.doi.org/10.1007/s00580-019-03028-4. Keith LS, Jones DE, Chou CH. (2002). Aluminum toxicokinetics regarding in-
Iijima Y, Bando M, Yamasawa H, Moriyama H, Takemura T, Niki T, Sugiyama Y. fant diet and vaccinations. Vaccine 20: 13–17.
(2017). A case of mixed dust pneumoconiosis with desquamative intersti- Kell BD. (2009). Iron behaving badly: inappropriate iron chelation as a major
tial-like reaction in an aluminum welder. Respir Med Case Rep 20: 150–153. contributor to the aetiology of vascular and other progressive inflamma-
tory and degenerative diseases. BMC Med Genomics 2: 1–79.
Issa AM, Salim MS, Zidan H, Mohamed AF, Farrag AH. (2014). Evaluation of the
effects of aluminum phosphate and calcium phosphate nanoparticles as Khalaf AA, Morgan AM, Mekawy MM, Ali MF. (2007). Developmental toxicity
adjuvants in vaccinated mice. Int J Chem Eng Appl 5: 367–373. evaluation of oral aluminum in rats. J Egypt Soc Toxicol 37: 11–26.
Jang JH, Surh YJ. (2002). β-Amyloid induces oxidative DNA damage and cell Khattab HAH, Abdallah IZA, Kamel GM. (2010). Grape seed extract alleviate
death through activation of c-Jun N terminal kinase. Ann N Y Acad Sci 973: reproductive toxicity caused by aluminium chloride in male rats. J Ameri-
228–236. can Sci 6: 1200–1209.
Jangra A, Kasbe P, Pandey SN, Dwivedi S, Gurjar SS, Kwatra M, Mishra M, Venu Khosla SN, Nand N, Khosla P. (1988). Aluminum phosphide poisoning. J Trop
AK, Sulakhiya K, Gogoi R, Sarma N, Bezbaruah BK, Lahkar M. (2015). Hesper- Med Hyg 91: 196–198.
idin and silibinin ameliorate aluminum-induced neurotoxicity: modulation Kihira T, Yoshida S, Yase Y, Ono S, Kondo T. (2002). Chronic low- Ca/Mg high–
of antioxidants and inflammatory cytokines level in mice hippocampus. Al diet induces neuronal loss. Neuropathology 22: 171–179.
Biol Trace Elem Res 168: 462–471. King SW, Savory J, Wills MR, Gitelman HJ. (1981). The clinical biochemistry of
Järup L. (2003). Hazards of heavy metal contamination. British Med Bull 68: aluminum. Crit Rev Clin Lab Sci 14: 1–20.
167–82. Klaassen CD. (1990). Heavy metals and heavy-metal antagonists, in Goodman
Jederlinic PJ, Abraham JL, Churg A, Himmelstein JS, Epler GR, Gaensler EA. and Gilman’s: The Pharmacological Basis of Therapeutics (Gilma AG, Rall TW,
(1990). Pulmonary fibrosis in aluminum oxide workers. Investigation of Nies AS, Taylor P eds), pp 1592–1614, Pergamon Press, New York.
nine workers with pathologic examination and microanalysis in three of Klein GL. (2019). Aluminum toxicity to bone: A multisystem effect. Osteoporos
them. Am Rev Respir Dis 142(5): 1179–1184. Sarcopenia 5(1): 2–5.
Jeffery EH, Abreo K, Burgess E, Cannata J, Greger JL. (1996). Systemic alumi- Klein JP, Mold M, Mery L, Cottier M, Exley C. (2014). Aluminum content of hu-
num toxicity: effect on bone haematopoietic tissue and kidney. J Toxicol man semen: implications for semen quality. Reprod Toxicol 50: 43–48.
Environ Health 46(6): 649–665.
Koltz K, Weistenhöfer W, Neff F, Hartwig A, van Thriel C, Drexler H. (2017). The
Jones KC, Bennet BG. (1986). Exposure of man to environmental aluminium- health effects of aluminum exposure. Dtsch Arztebl Int 114(39): 653–659.
an exposure commitment assessment. Sci Total Environ 52: 65–82.
Konda VR, Eerike M, Chary RP, Arunachalam R, Yeddula VR, Meti V, Devi TS.
Jones K, Linhart C, Haekin C, Exley C. (2017). Urinary excretion of aluminium (2017). Effect of aluminum chloride on blood glucose level and lipid profile
and silicon in secondary progressive multiple sclerosis. EBioMedicine 26: in normal, diabetic and treated diabetic rats. Indian J Pharmacol 49: 357–365.
60–67.
Kongerud J, Søyseth V. (2014). Respiratory disorders in aluminum smelter
Johnson VJ, Sharma RP. (2003). Aluminum disrupts the pro-inflammatory cy- workers. J Occup Environ Med 56(5 suppl): S60–70.
tokine/neurotrophin balance in primary brain rotation-mediated aggre-
Konishi Y, Yagyu K, Kinebuchi H, Salto N, Yamaguchi T, Ohtsuki Y. (1996).
gate cultures: possible role in neurodegeneration. Neurotoxicology 24(2): Chronic effect of aluminum ingestion on bone in calcium-deficient rats.
261–268. Basic Clin Pharmacol Toxicol 78: 429–434.
Jouhanneau P, Raisbeck GM, Yiou F, Lacour B, Banide H, Drueke TB. (1997). Kowalczyk E, Kopff A, Kędziora J, Błaszczyk J, Kopff M, Niedworok J,
Gastrointestinal absorption, tissue retention, and urinary excretion of di- Fijałkowski P. (2004). Effect of long-term aluminium chloride intoxication
etary aluminum determined by using 26Al. Clin Chem 43: 1023–1028. on selected biochemical parameters and oxidative-antioxidative balance
Julka D, Gill KD. (1996). Altered calcium homeostasis: a possible mechanism in experimental animals. Pol J Environ Stud 13: 41–43.
of aluminium-induced neurotoxicity. Biochem Biophys Acta 1315: 47–54. Krewski D, Yokel RA, Nieboer E, Borchelt D, Cohen J, Harry J, Kacew S, Lind-
Kaiser L,Schwartz KA, Burnatowska-Hledin MA. (1984). Microcytic anemia say J, Mahfouz AM, Rondeau V. (2007). Human health risk assessment for
secondary to intraperitoneal aluminum in normal and uremic rats. Kidney aluminium, aluminium oxide, and aluminium hydroxide. J Toxicol Environ
Int 26: 269–274. Health 10: 251–269.

Copyright © 2019 SETOX & Institute of Experimental Pharmacology and Toxicology, CEM SASc.
66 Aluminium toxicosis
Ikechukwu Onyebuchi Igbokwe, Ephraim Igwenagu, Nanacha Afifi Igbokwe

Kücük M, Kalayci RB, Cevik A, Elmas I, Kaya M. (2001). Effect of aluminum on Mailloux RJ, Lemire J, Appanna VD. (2011). Hepatic response to aluminum
the blood-brain barrier permeability in acute and chronically hyperglycae- toxicity: dyslipidemia and liver diseases. Exp Cell Res 317: 2231–2238.
mic rats. Biol Trace Elem Res 80 (2): 181–189. Maitani T, Kubota H, Hori N. (1994). Distribution and urinary excretion of alu-
Kumar V, Gill KD. (2014). Oxidative stress and mitochondrial dysfunction in al- minum injected with several organic acids into mice: Relationship with
uminium neurotoxicity and its amelioration. Neurotoxicology 41: 154–166. chemical state in serum studied by the HPLC-ICP method. J Appl Toxicol 14:
Lantzy RJ, MacKenzie FT. (1979). Atmospheric trace metals: global cycles and 257–261.
assessment of man’s impact. Geochim Cosmochim Acta 43: 511–525. Malakoff D. (2000). Aluminum is put on trial as a vaccine booster. Science 288:
Lee REJ, Von Lehmden DJ. (1973). Trace metal pollution in the environment. J 1323–1234.
Air Pollut Control Assoc 23: 853–857. Malekshah AK, Torabizadeh Z, Naghshwar F. (2005). Developmental toxicity
Lefcourt AM, Mesinger JJ. (2001). Effect of adding alum or zeolite to dairy of aluminum from high doses of AlCl3 in mice. J Appl Res 5(4): 575–579.
slurry on ammonia volatilization and chemical composition. J Dairy Sci 84: Malluche HH, Smith AJ, Abreo K, Faugere MC. (1984). The use of deferox-
1814–1821. arnme in the management of aluminum accumulation in bone in patients
Lerner A. (2007). Aluminum is a potential envirnmental factor for Crohn’s dis- with renal failure. N Engl J Med 311: 140–144.
ease induction: extended hypothsesis. Ann N Y Acad Sci 1107: 329–345. Manello F, Ligi D, Canale M. (2013). Aluminium, carbonyls and cytokines in
human nipple aspirate fluids: possible relationship between inflammation,
Levine S, Saltzman A, Drakontides AB. (1992). Parenteral aluminum com-
oxidative stress and breast cancer microenvirnment. J Inorg Biochem 128:
pounds produce a local toxic myopathy in rats: importance of the anion.
250–256.
Toxicol Pathol 20(3–1): 405–415.
Marítin-Llorens M, Jurado J, Hernández F, Avila J. (2014). GSK-3β, a pivotal ki-
Levine SN, Sonnier GB, Abreo K. (1990). Effects of diabetes mellitus and alu-
nase in Alzheimer’s disease. Front Mol Neurosci 7: 46.
minum toxicity on myocardial calcium transport. Toxicology 65: 137–148.
Martin RB. (1992). Aluminum speciation in biology. Ciba Found Symp 16: 95–
Lewis RJ. (2001). Hawley’s Condensed Chemical Dictionary, 14th edn, pp. 39–
125.
46, Wiley-Interscience, New Jersey, USA.
Martin RB, Savory J, Brown S, Bertholf RL, Wills MR. (1987). Transferrin binding
Li P, Luo W, Zhang H, Zheng X, Liu C, Ouyang H. (2016). Effects of aluminum
to Al and Fe. Clin Chem 33: 405–407.
exposure on bone stimulatory growth factors in rats. Biol Trace Elem Res
172(1): 166–171. Martinez CS, Escobar AG, Uranga-Ocio JA, Precanba FM, Vassallo DV, Exley C,
Miguel M, Wiggers GA. (2017). Aluminum exposure for 60 days at human
Li X, Han Y, Guan Y, Zhang L, Bai C, Li Y. (2012). Aluminum induces osteoblast
dietary levels impairs spermatogenesis and sperm quality in rats. Reprod
apoptosis through oxidative stress-mediated JNK signaling pathway. Biol Toxicol 73: 128–141.
Trace Elem Res 150(1–3): 502–508.
Martyn CN, Barker DJ, Osmond C, Harris EC, Edwardson JA, Lacey RF. (1989).
Li Y, Liu J, Cao Z. (2015). Effect of sub-chronic aluminum exposure on renal Geographical relation between Alzheimer’s disease and aluminum in
structure in rats. J Northeast Agric Univ 22(2): 47–51. drinking water. Lancet 1: 59–62.
Liaquat L, Sadir S, Batool Z, Tabassum S, Shahzad S, Afzal A, Haider S. (2019). Mathiyazahan DB, Thenmozhi AJ, Manivasagam T. (2015). Protective effect of
Acute aluminum chloride toxicity revisited: study on DNA damage and his- black tea extract against aluminium chloride-induced Alzheimer’s disease
topathological, biochemical and neurochemical alterations in rat brain. in rats. A behavioural, biochemical and molecular approach. J Funct Foods
Life Sci 217: 202–211. 16: 423–435.
Lidsky TI. (2014). Is the aluminum hypothesis dead? J Occup Environ Med 56(5 Maya S, Prakash T, Madhu KD, Goli D. (2016). Multifaceted effects of alumin-
suppl): S73–S79. ium in neurodegenerative diseases: a review. Biomed Pharmacother 83:
Lin CY, Hsiao WC, Huang CJ, Kao CF, Hsu GS. (2013). Heme oxygenase-1 induc- 746–754.
tion by ROS-JNK pathway plays a role in aluminum-induced anemia. J Inorg Mayor GH, Lohr TO, Sanchez TV. (1985). Aluminum metabolism and toxicity in
Chem 128: 221–228. renal failure: A review. J Environ Pathol Toxicol Oncol 6: 43–50.
Lin JL, Yang YJ, Yang SS, Leu ML. (1997). Aluminum utensils contribute to alu- Mehrpour O, Alfred S, Shadnia S, Keyler DE, Soltaninejad K, Chalaki N, Sed-
minum accumulation in patients with renal disease. Am J Kidney Dis 30: aghat M. (2008). Hyperglycemia in acute aluminum phosphide poisoning
653–665. as a potential prognostic factor. Hum Exp Toxicol 27: 591–595.
Linhart C, Talasz H, Morandi EM, Exley C, Lindner HH, Taucher S, Egle D, Hu- Meliker JR, Wahi RL, Cameron LL, Nriagu JO. (2007). Arsenic in drinking wa-
balek M, Concin N, Ulmer H. (2017). Use of underarm cosmetic products in ter and cerebrovascular disease, diabetes mellitus, and kidney disease in
relation to risk of breast cancer: a case-control study. EBioMedicine 21: 78– Michigan: a standardized mortality ratio analysis. Environ Health 6: 1–11.
85.
Menke A, Casagrande S, Geiss L, Cowie CC. (2015). Prevalence of and trends
Lione A. (1983). The prophylactic reduction of aluminum intake. Food Chem in diabetes among adults in the United States, 1988–2012. J Am Med Assoc
Toxicol 21: 103–109. 314(10): 1021–1029.
Lione A. (1985). Aluminum toxicity and the aluminum-containing medica- Mestaghanmi H, El Amrani S, Dauca M, Saile R. (2003). Effect of aluminium
tions. Pharmacol Ther 29: 255–285. chloride on pregnant rats and their offspring. Sciences et Techniques de
Liukkonen-Lilja H, Piepponen S. (1992). Leaching of aluminum from alumi- l’Animal de Laboratoire 28(1): 43–51.
num dishes and packages. Food Addit Contam 9: 213–223. Miller RR, Churg AM, Hutcheon M, Lom S. (1984). Pulmonary aveolar pro-
Lukiw WJ, LeBlanc HJ, Carver LA, McLachlan DR, Bazan NG. (1998). Run-on teinosis and aluminum dust exposure. Am Rev Respir Dis 130(2): 312–315.
gene transcription in human neocortical nuclei. Inhibition by nanomolar Miller ZN. (2016). Aluminum in childhood vaccines is unsafe. J Am Phys Surg
aluminum and implications for neurodegenerative disease. J Mol Neurosci 21(4): 109–117.
11: 67–78.
Milnerowicz H, Ściskalska M, Dul M. (2015). Pro-inflammatory effects of met-
Lukiw WJ, Percy ME, Kruck TP. (2005). Nanomolar aluminum induces pro-in- als in persons and animals exposed to tobacco smoke. J Trace Elem Med Biol
flammatory and pro-apoptotic gene expression in human brain cells in pri- 29: 1–10.
mary culture. J Inorg Biochem 99(9): 1895–1898. Mirhashemi SM, Aarabi MH. (2011). To study various concentrations of mag-
Lukyanenko LM, Skarabahatava AS, Slobozhanina EI, Kovaliova SA, Falcioni nesium and aluminium on amylin hormone conformation. Pak J Biol Sci
ML. (2013). In vitro effect of AlCl3 on human erythrocytes: changes in mem- 14(11): 653–657.
brane morphology and functionality. J Trace Elem Med Biol 27(2): 160–167. Mirhashemi SM, Shahabaddin M-E. (2011). Evaluation of aluminium, manga-
Lyons-Weiler J, Ricketson R. (2018). Reconsideration of the immunotherapeu- nese, copper and selenium effects on human islets amyloid polypeptide
tic pediatric safe dose levels of aluminum. J Trace Elem Med Biol 48: 67–73. hormone aggregation. Pak J Biol Sci 14(4): 288–292.
Mahieu S, Contini MC, Gonzalez M, Millen N, Elias MM. (2000). Aluminum tox- Mirza A, King A, Troakes C, Exley C. (2017). Aluminium in brain tissue in famil-
icity. Hematological effects. Toxicol Lett 111(3): 235–242. ial Alzheimer’s disease. J Trace Elem Med Biol 40: 30–36.
Mailloux RJ, Hamel R, Appanna VD. (2006). Aluminum toxicity elicits a dys- Miska-Schramm A, Kapusta A, Kruczek M. (2017). The effect of aluminum ex-
functional TCA cycle and succinate accumulation in hepatocytes. J Bio- posure on reproductive ability of the bank vole (Myodes glareolus). Biol
chem Mol Toxicol 20: 198–208. Trace Elem Res 177(1): 97–106.

ISSN: 1337-6853 (print version) | 1337-9569 (electronic version)


Interdisciplinary Toxicology. 2019; Vol. 12(2): 45–70 67
Full-text also available online on PubMed Central

Mitkus RJ, King DB, Hess MA, Forshee RA, Walderhaug MO. (2011). Updated Nikolov IG, Joki N, Vicca S, Patey N, Auchere D, Benchitrit J. (2010). Tissue ac-
aluminum pharmacokinetics following infant exposures through diet and cumulation of lanthanum as compared to aluminum in rats with chronic
vaccination. Vaccine 29(51): 9538–9543. renal failure: possible harmful effects after long-term exposure. Nephron
Mittal K, Katare DP. (2016). Shared links between type 2 diabetes mellitus and Exp Nephrol 115: 112–121.
Alzheimer’s disease: A review. Diabetes Metab Syndr 10(2 Suppl 1): S144– Nordal KP, Dahl E, Albrechtsen D, Halse J, Leivestad T, Tretli S, Flatmark A.
S149. (1988). Aluminum accumulation and immunosuppressive effect in recipi-
ents of kidney transplants. Br Med J 287: 1581–1582.
Miu AC, Benga O. (2006). Aluminum and Alzheimer’s disease: a new look. J Al-
zheimers Dis 10(2–3): 179–201. Obukhova T, Budkar’ LN, Tereshina LG, Karpova EA. (2015). [Dissociation of
disorders of carbohydrate and lipid metabolism in aluminum industry
Moghadamnia AA. (2012). An update on toxicology of aluminum phosphide. workers according to medical examination data] in Russian. Gig Sanit 94
DARU J Pharm Sci 20: 1–8. (2): 67–69.
Mohammed A, Mayyas I, Elbetieha A, Shoter A, Khamas W, Alnasser Z. (2008). Ogborn MR, Dorcas VC, Crocker JF. (1991). Deferoxamine and aluminum
Toxicity evaluation of aluminium chloride on adult female mice. J Anim Vet clearance in pediatric hemodialysis patients. Pediatr Nephrol 5: 62–64.
Adv 7(5): 552–556.
Ohsaka Y, Nomura Y. (2016). Rat white adipocytes activate p85/p110 P13K and
Mold M, Umar D, King A, Exley C. (2018). Aluminium in brain tissue in autism. induce PM GLUT4 response to adrenoceptor agonists or aluminum fluo-
J Trace Elem Med Biol 46: 76–82. ride. Physiol Int 103(1): 35–48.
Moore PA, Daniel TC,Edwards DR. (1999). Reducing phosphorus runoff and Omar HM, Khadiga AH, Abd-Elghaffar SK, Ahmed EA. (2003). Aluminium tox-
improving poultry production with alum. Poult Sci 78: 692–698. icity in rats: the role of tannic acid as antioxidant. Assiut Univ Bull Environ
Moore PA, Daniel TC,Edwards DR. (2000). Reducing phosphorus runoff and Res 6: 1–14.
inhibiting ammonia loss from poultry manure with aluminum sulfate. J En- Ondov JM, Zoller WH, Gordon GE. (1982). Trace element emissions of aerosols
viron Qual 29: 37–49. from motor vehicles. Environ Sci Technol 16: 318–328.
Morris G, Puri BK, Frye RE. (2017). The putative role of environmental alumin- Oneda S, Takasaki T, Kuriwaki K, Ohi Y, Umekita Y, Hatanaka S, Fujiyoshi T, Yo-
ium in the development of chronic neuropathology in adults and children. shida A, Yoshida H. (1994). Chronic toxicity and tumorigenicity study of alu-
How strong is the evidence and what could be the mechanism involved? minum potassium sulfate in B6C3F1 mice. In Vivo 8: 271–278.
Metab Brain Dis 32(5): 1335–1355. Orihuela D. (2007). Effect of aluminium on duodenal calcium transport in
Morrissey J, Rothstein M, Mayor G, Slatopolsky E. (1983). Suppresion of para- pregnant and lactating rats treated with bromocriptine. J Inorg Biochem
thyroid hormone secretion by aluminum. Kidney Int 23(5): 699–704. 101(9): 1270–1274.
Mouro VGS, Menezes TP, Lima GDA, Domingues RR, Souza AC, Oiveira JA, Orihuela D. (2011). Aluminium effects on thyroid gland function: iodide up-
Matta SLP, Machado-Neves M. (2017). How bad is aluminum exposure to take, hormone biosynthesis and secretion. J Inorg Biochem 105(11): 1464–
reproductive parameters? Biol Trace Elem Res Sept 8. Doi: 10.1007/s12011- 1468.
017-1139-3. Osama A, Fatma A, Mohamed E, Huda S. (2014). Studies on the protective ef-
Mujika JI, Torre GD, Formoso E, Grande-Aztatzi R, Grabowski SJ, Exley C, Lo- fects of ginger extract and in combination with ascorbic acid against alu-
pex X. (2018). Aluminum’s preferential binding site in proteins: sidechain minum toxicity induced hematological disorders, oxidative stress and hep-
of amino acids versus backbone interactions. J Inorg Biochem 181: 111–116. atorenal damage in rats. Ann Vet Anim Sci 1: 136–150.
Osman HM, Shayoub ME, Babiker EM, Osman B, Elhassan AM. (2012). Effect of
Muselin F, Cristina RT, Iqna V, Dumitrescu E, Brezovan D, Trif A. (2016). The
ethanolic leaf extract of Moringa oleifera on aluminum-induced anemia in
consequences of aluminium intake on reproductive function in male rats: a
white albino rats. Jordan J Biol Sci 5: 255–260.
three-generation study. Turk J Med Sci 46: 1240–1248.
Oteiza PI. (1994). A mechanism for the stimulatory effect of aluminum on
Nam SM, Kim JW, Yoo DY, Kim W, Jung HY, Hwang IK, Seong JK, Yoon YS.
iron-induced lipid peroxidation. Arch Biochem Biophys 308: 374–379.
(2014). Additive or synergistic effects of aluminum on the reduction of neu-
ral stem cells, cell proliferation and neuroblast differentiation in the den- Oteiza PI, Fraga C, Keen CL. (1993a). Aluminum has both oxidant and antioxi-
tate gyrus of high-fat diet-fed mice. Biol Trace Elem Res 157 (1): 51–59. dant effects in mouse brain membranes. Arch Biochem Biophys 300: 517–521.
Nam SM, Kim JW, Yoo DY, Jung HY, Choi JH, Hwang IK, Seong JK, Yoon YS. Oteiza PI, Keen CL, Han B, Golub MS. (1993b). Aluminum accumulation and
(2016). Reduction of adult hippocampal neurogenesis is amplified by alu- neurotoxicity in Swiss-Webster mice after long-term dietary exposure to
aluminum and citrate. Metabolism 42: 1296–1300.
minium exposure in a model of type 2 diabetes. Journal of Veterinary Sci-
ence 17(1): 13–20. Owen LM, Crews HM, Bishop NJ, Massey RC. (1994). Aluminum uptake from
some foods by guinea pigs and the characterization of aluminum in in vivo
Nasu T, Suzuki N. (1998). Effect of aluminum ions on K+-induced contraction
intestinal digesta by SEC-ICP-MS. Food Chem Toxicol 32: 697–705.
in ileal longitudinal smooth muscle. Comp Biochem Physiol C Pharmacol
Toxicol Endocrinol 120(1): 137–143. Oztürk B, Ozdemir S. (2015). Effects of aluminum chloride on some trace el-
ements and erythrocyte osmotic fragility in rats. Toxicol Ind Health 31(12):
Nayak P. (2002). Aluminum: impacts and disease. Environ Res 89: 101–115. 1069–1077.
Navas-Acien A, Silbergeld EK, Pastor-Barriuso R, Guallar E. (2009). Rejoinder: Pandey G, Jain GC. (2013). A review on toxic effects of aluminium exposure
arsenic exposure and prevalence of type 2 diabetes: updated findings from on male reproductive system and probable mechanisms of toxicity. Intl J
the National Health Nutrition and Examination Survey, 2003–2006. Epide- Toxicol Appl Pharmacol 3(3): 48–57.
miology 20: 816–820.
Pappas RS. (2011). Toxic elements in tobacco and cigarette smoke: inflamma-
Neiva TJC, Fries DM, Monteiro HP, D’Amico EA, Chamone DAF. (1997). Alumi- tion and sensitization. Metallomics 3(11): 1181–1198.
num induces lipid peroxidation and aggregation of human blood plate-
Pennington JA. (1988). Aluminum content of foods and diets. Food Addit Con-
lets. Braz J Med Biol Res 30: 599–604.
tam 5: 161–232.
Nelson WO, Campbell PGC. (1991). The effects of acidification on the geo- Pennington JA,Schoen SA. (1995). Estimates of dietary exposure to alumin-
chemistry of Al, Cd, Pb, and Hg in freshwater environments: a literature re- ium. Food Addit Contam 12: 119–128.
view. Environ Pollut 71: 91–130.
Perez G, Garbossa G, Nesse A. (2001). Disturbance of cellular iron uptake and
Netterlid E, Hindsén M, Siemund I, Björk J, Werner S, Jacobsson H, Güner N, utilization by aluminum. J Inorg Biochem 87: 21–27.
Bruze M. (2013). Does allergen-specific immunotherapy induce contact al-
Pettersen JC, Hackett DS, Zwicker GM,Sprague GL. (1990). Twenty-six week
lergy to aluminim? Acta Derm Venereol 93: 50–56.
toxicity study with KASAL (basic sodium aluminum phosphate) in beagle
Newairy AS, Salama AF, Hussien HM, Yousef MI. (2009). Propolis alleviates dogs. Environ Geochem Health 12: 121–123.
aluminum-induced lipid peroxidation and biochemical parameters in male
Pineau A, Fauconneau B, Sappino A-P, Deloncle R, Guillard O. (2014). If ex-
rats. Food Chem Toxicol 47: 1093–1098.
posure to aluminium in antiperspirants presents health risk, its content
Ng M. (2004). Role of hydrogen peroxide in the aetiology of Alzheimer’s dis- should be reduced. J Trace Elem Med Biol 28(2): 147–150.
ease: Implications for treatment. Drugs Aging 21: 81–100. Pivnick EK, Kerr NC, Kaufman RA, Jones DP, Chesney RW. (1995). Rickets sec-
Niemoeller OM, Kiedaisch V, Dreischer P, Wieder T, Lang F. (2006). Stimula- ondary to phosphate depletion: a sequela of antacid use in infancy. Clin Pe-
tion of eryptosis by aluminum ions. Toxicol Appl Pharmacol 217(2): 168–175. diatr 34: 73–78.

Copyright © 2019 SETOX & Institute of Experimental Pharmacology and Toxicology, CEM SASc.
68 Aluminium toxicosis
Ikechukwu Onyebuchi Igbokwe, Ephraim Igwenagu, Nanacha Afifi Igbokwe

Pogue AI, Jaber V, Zhao Y, Lukiw WJ. (2017). Systemic inflammation in Schroeder HA, Mitchener M. (1975b). Life-term effects of mercury, methyl-
C57BL/6J mice receiving dietary aluminum sulfate; up-regulation of the mercury, and nine other trace metals on mice. J Nutr 105: 452–458.
pro-inflammatory cytokines IL-6 and TNFα, C-reactive protein (CRP) and Sedman AB, Klein GL, Merritt RJ, Weber KO, Gill WL, Anand H, Alfrey AC.
miRNA-146a in blood serum.  J Alzheimers Dis Parkinsonism  7(6): 403. doi: (1985). Evidence of aluminum loading in infants receiving intravenous
10.4172/2161-0460.1000403. therapy. N Engl J Med 312(21): 1337–1343.
Pogue Ai, Lukiw WJ. (2016). Aluminium, the genetic apparatus of human CNS Sepe A, Costantini S, Ciaralli L, Ciprotti M, Giordano R. (2001). Evaluation of
and Alzheimer’s disease. Morphologie 100(329): 56–64. aluminium concentrations in samples of chocolate and beverages by elec-
Pratico D, Uryu K, Sung S, Tang S, Trojanowski JQ, Lee VM. (2002). Alumi- trothermal atomic absorption spectrometry. Food Addit Contam 18: 788–
num modulates brain amyloidosis through oxidative stress APP transgenic 796.
mice. FASEB J 16: 1138–1140. Serdar MA, Bakir F, Hasimi A, Celik T, Akin O, Kenar L, Aykut O, Yildirimkaya M.
Priest ND, Talbot RJ, Austin JG, Day JP, King SJ, Fifield K, Cresswell RG. (1996). (2009). Trace and toxic element patterns in nonsmoker patients with non-
The bioavailability of 26Al-labelled aluminium citrate and aluminium hy- insulin-dependent diabetes mellitus, impaired glucose tolerance, and fast-
droxide in volunteers. Biometals 9: 221–228. ing glucose. Int J Diabetes Dev Ctries 29: 35–40.
Pun KK, Ho PWM, Lau P. (1990). Effect of aluminum on the parathyroid hor- Shadnia S, Mehrpour O, Abdollahi M. (2008). Unintentional poisoning by
mone receptors of bone and kidney. Kidney Int 37(1): 72–78. phosphine released from aluminum phosphide. Hum Exp Toxicol 27: 87–89.
Que Hee SS, Finelli VN, Fricke FL,Wolnik KA. (1982). Metal content of stack Shahraki MR, Mony EYP, Asl SZ, Sarkaki AR, Shahraki AR. (2008). Effects of alu-
emissions, coal and fly ash from some eastern and western power plants in minium chloride injection in lateral ventricles on serum gonadothropines,
the U.S.A. as obtained by ICP-AES. Int J Environ Anal Chem 13: 1–18. testosterone and spermatogenesis in rats. J Med Sci 8(4): 410–414.
Qureshi N, Malmberg RH. (1985). Reducing aluminum residuals in finished Sharma DR, Wani WY, Sunkaria A, Kandimalla RJ, Sharma RK, Verma D, Bal A,
water. J Am Water Works Assoc 77: 1–18. Gill KD. (2016). Quercetin attenuates neuronal death against aluminum-in-
Ranau R, Oehlenschlager J, Steinhart H. (2001). Aluminium levels of fish fillets duced neurodegeneration in rat hippocampus. Neuroscience 324: 163–176.
baked and grilled in aluminium foil. Food Chem 73: 1–6. Shati AA, Alamri SA. (2010). Role of saffron (Crocus sativus L.) and honey syrup
Ranjbar A, Khani-Jazani R, Sedighi A, Jalali-Mashayekhi F, Ghazi-Khansari M, on aluminum-induced hepatotoxicity. Saudi Med J 31: 1106–1113.
Abdollahi M. (2008). Alteration of body total antioxidant capacity and thiol Shaw CA, Li D, Tomljenovic L. (2014). Are there negative CNS impacts of alu-
molecules in human chronic exposure to aluminum. Toxicol Environ Chem minum adjuvants used in vaccines and immunotherapy. Immunotherapy
90: 707–713. 6(10): 1055–1077.
Ravi SK, Ramesh BN, Mundugaru R, Vincent B. (2018). Multiple pharmacologi- She Y, Wang N, Chen C, Zhu Y, Xia S, Hu C, Li Y. (2012). Effects of aluminum
cal activities of Caesalpinia crista against aluminium-induced neurodegen- on immune functions of cultured splenic T and B lymphocytes in rats. Biol
eration in rats: Relevance for Alzheimer’s disease. Environ Toxicol Pharmacol Trace Elem Res 147(1–3): 246–250.
58: 202–211.
Sherrard DJ, Ott SM, Andress DL. (1985). Pseudohyperparathyroidism syn-
Razniewska G, Trzcinka-Ochocka M. (2003). ET-AAS as a method for determi- drome associated with aluminum intoxication in patients with renal fail-
nation of aluminum in blood serum and urine. Chem Anal 48: 107–113. ure. Am J Med 79(1): 127–130.
Reinke CM, Breitkreutz J, Leuenberger H. (2003). Aluminum in over the-coun- Shirley DG, Lote CJ. (2005). Renal Handling of Aluminium. Nephron Physiol
ter drugs: risks outweigh benefits. Drug Saf 26: 1011–1025. 100: 99–103.
Reto M, Figueira ME, Filipe HM, Almeida CM. ( 2007). Chemical composition Singh T, Goel RK. (2015). Neuroprotective effect of Allium cepa L. in alumin-
of green tea (Camellia sinensis) infusions commercialized in Portugal. Plant ium chloride induced neurotoxicity. NeuroToxicology 49: 1–7.
Foods Hum Nutr 62: 139–144.
Skarabahatava AS, Lukyanenko LM, Slobozhanina EI, Falcioni ML, Orlando P,
Reza SM, Palan MJ. (2006). Effect of aluminium on testosterone hormones in Silvestri S, Tiano L, Falcioni G. (2015). Plasma and mitochondrial membrane
male rat. J Med Sci 6(2): 296–299. perturbation induced by aluminum in human peripheral blood lympho-
Rigolet M, Aouizerate J, Couette M, Ragunathan-Thangarajah N, Aoun-Se- cytes. J Trace Elem Med Biol 31: 37–44.
baiti M, Gherardi RK, Cadusseau C, Authier FJ. (2014). Clinical features in Song M, Huo H, Cao Z, Han Y, Gao L. (2017). Aluminum trichloride inhibits the
patients with long-lasting macrophagic myofasciitis. Front Neurol doi: rat osteoblast mineralization in vitro. Biol Trace Elem Res 175(1): 186–193.
10.3389/fneur.2014.00230.
Soni MG, White SM, Flamm WG, Burdock GA. (2001). Safety evaluation of di-
Rim K. (2007). Aluminum leaching using chelating agent as a composition etary aluminum. Regul Toxicol Pharmacol 33: 66–79.
food. Food Chem Toxicol 45: 1688–1693.
Sorenson JRJ, Campbell IR, Tepper LB,Lingg RD. (1974). Aluminum in the en-
Rodríguez J, Mandlunis PM. (2018). A review of metal exposure and its effects
vironment and human health. Environ Health Perspect 8: 3–95.
on bone health. J Toxicol 2018, Article ID 4854152, 11 pages.
Sorgdrager B, Looff AJ, Monchy JG, Pal TM, Dubois AE, Rijcken B. (1998). Oc-
Rosseland BO, Eidhuset TD, Staurnes M. (1990). Environmental effects of alu-
currence of occupational asthma in aluminum potroom workers in relation
minum. Environ Geochem Health 12: 17–27.
to preventative measures. Int Arch Occup Environ Health 71: 53–59.
Sadhana S. (2011). S-Allyl-Cysteines reduce amelioration of aluminum in-
Stahl T, Taschan H, Brunn H. (2011). Aluminium content of selected foods and
duced toxicity in rats. Am J Biochem Biotechnol 7: 74–83.
food products. Environ Sci Eur 23(37): 1–11.
Saiyed SM, Yokel RA. (2005). Aluminum content of some foods and food
Steinhausen C, Kislinger G, Winklhofer C. (2004). Investigation of the alumi-
products in the USA, with aluminum food additives. Food Addit Contam 22:
num biokinetics in humans: a 26Al tracer study. Food Chem Toxicol 42: 363–
234–244.
371.
Sakajiri T, Yamamura T, Kikuchi T, Ichimura K, Sawada T, Yajima H. (2010). Ab-
Stone CJ, McLaurin DA, Steinhagen WH, Cavender FL, Haseman JK. (1979).
sence of binding between the human transferrin receptor and the trans-
Tissue deposition patterns after chronic inhalation exposures of rats and
ferrin complex of biological toxic trace element, aluminum, because of an
incomplete open/closed form of the complex. Biol Trace Elem Res 136: 279– guinea pigs to aluminum chlorhydrate. Toxicol Appl Pharmacol 49: 71–76.
286. Sun H, Hu C, Jia L, Zhu Y, Zhao H, Shao B, Wang N, Zhang Z, Li Y. (2011). Effects
Sakamoto T, Saito H, Ishii K, Takahashi H, Tanabe S, Ogasawara Y. (2006). Alu- of aluminum exposure on serum sex hormones and androgen receptors
minum inhibits proteolytic degradation of amyloid beta peptide by ca- expression in male rats. Biol Trace Elem Res 144(1–3): 1050–1058.
thepsin D: a potential link between aluminum accumulation and neuritic Sun X, Cao Z, Zhang Q, Han L, Li Y. (2016a). Aluminum chloride inhibits osteo-
plaque deposition. FEBS Lett 580: 6543–6549. blast mineralization via TGF-β1/Smad signaling pathway. Chem Biol Interact
Savory J, Exley C, Forbes WF, Huang Y, Joshi JG, Kruck T, McLachlan DR, 244: 9–15.
Wakayama I. (1996). Can the controversy of the role of aluminum in Al- Sun X, Cao Z, Zhang Q, Liu S, Xu F, Che J, Zhu Y, Li, Y, Pan C, Liang W. (2015).
zheimer’s be resolved? What are the suggested approaches to this contro- Aluminum trichloride impairs bone and downregulates Wnt/β-catenin sig-
versy and methodological issues to be considered? J Toxicol Environ Health naling pathway in young growing rats. Food Chem Toxicol 86: 154–162.
48: 615–635. Sun X, Liu J, Zhuang C, Yang X, Han Y, Shao B, Song M, Li Y, Zhu Y. (2016b).
Schroeder HA, Mitchener M. (1975a). Life-term studies in rats: effects of alu- Aluminum trichloride induces bone impairment through TGF-β1/Smad sig-
minum, barium, beryllium and tungsten. J Nutr 105: 421–427. naling pathway. Toxicology 371: 49–57.

ISSN: 1337-6853 (print version) | 1337-9569 (electronic version)


Interdisciplinary Toxicology. 2019; Vol. 12(2): 45–70 69
Full-text also available online on PubMed Central

Sun X, Sun H, Yu K, Wang Z, Liu Y, Liu K, Zhu Y, Li Y. (2018). Aluminum chlo- Verma SK, Ahmad S, Shirazi N, Barthwal SP, Khurana D, Chugh M, Gambhir
ride causes the dysfunction of testes through inhibiting ATPase enzyme HS. (2007). Acute pancreatitis: a lesser-known complication of aluminum
activities and gonadotropin receptor expression in rats. Biol Trace Elem Res phosphide poisoning. Hum Exp Toxicol 26: 979–981.
183(2): 296–304. Vignal C, Desreumaux P, Body-Malapel M. (2016). Gut: An underestimated
Sun X, Wang H, Huang W, Yu H, Shen T, Song M, Han Y, Li Y, Zhu Y. (2017). Inhi- target organ for aluminium. Morphologie 100(329): 75–84.
bition of bone formation in rats by aluminum exposure via Wnt/β-catenin Vittori D, Garbossa G, Lafourcade C, Pérez G, Nesse A. (2002). Human ery-
pathway. Chemosphere 176: 1–7. throid cells are affected by aluminium. Alteration of membrane band 3
Sumathi T, Shobana C, Mahalakshmi V, Sureka R, Subathra M, Vishali A, Rekha protein. Biochim Biophys Acta Biomembr 1558(2): 142–150.
K. (2013). Oxidative stress in brains of male rats intoxicated with aluminium Vittori D, Nesse A, Pérez G, Garbossa G. (1999). Morphologic and functional
and neuromodulating effect of Celastrus paniculatus alcoholic seed ex- alterations of erythroid cells induced by long-term ingestion of aluminum.
tract. Asian J Pharm Clin Res 6: 80–90. J Inorg Biochem 76: 113–120.
Sushma NJ, Sivaiah U, Suraj NJ, Rao KJ. (2007). Aluminium acetate: role in oxi- Vittori D, Pregi N, Pérez G, Garbossa G, Nesse A. (2005). The distinct erythro-
dative metabolism of albino mice. Int Zool Res 3(1): 48–52. poietin functions that promote cell survival and proliferation are affected
Suwalsky M, Norris B, Villena F, Cuevas F, Sotomayor P, Zatta P. (2004). Alu- by aluminum exposure through mechanisms involving erythropoietin re-
minium fluoride affects the structure and functions of cell membrane. Toxi- ceptor. Biochim Biophys Acta 1743(1–2): 29–36.
cology 42(6): 925–933. Vorbrodt AW, Trowbridge RS, Drobrogowska DH. (1994). Cytochemical study
Taїr K, Kharoubi O, Taїr OA, Hellal N, Benyettou I, Aoues A. (2016). Aluminium- of the effect of aluminium on cultured brain microvascular endothelial
induced acute neurotoxicity in rats: Treatment with aqueous extract of Ar- cells. Histochem J 26(2): 119–126.
throphytum (Hammada scoparia). J Acute Dis 5(6): 470–482. Vota DM, Crisp RL, Nesse AB, Vittori DC. (2012). Oxidative stress due to alumi-
Taiwo OA. (2014). Diffuse parenchymal diseases associated with aluminium num exposure induces eryptosis which is prevented by erythropoietin. J
use and primary aluminum production. J Occup Environ Med 55(5 suppl): Cell Biochem 113(5): 1581–1589.
S71–S72. Wang N, She Y, Zhu Y, Zhao H, Shao B, Sun H, Hu C, Li Y. (2012). Effects of sub-
Taiwo OA, Sircar KD, Slade MD, Cantley LF, Vegso SJ, Rabinowitz PM, Fiellin chronic aluminum exposure on the reproductive function of female rats.
Biol Trace Elem Res 145(3): 382–387.
MG, Cullen MR. (2006). Incidence of asthma among aluminum workers. J
Occup Environ Med 48(3): 275–282. Wang X, Xi Y, Zeng X, Zhao H, Cao J, Jiang W. (2018). Effect of chlorogenic acid
against aluminium neurotoxicity in ICR mice through chelation and anti-
Tanabe K, Liu Z, Patel S, Doble BW, Li L, Cras-Méneur C, Martinez SC, Well-
oxidant actions. J Funct Foods 40: 365–376.
ing CM, White MF, Bernal-Mirzrachi E, Woodgett JR, Permutt MA. (2008).
Genetic deficiency of glycogen synthase kinase-3β corrects diabetes in Wang Z, Wei X, Yang J, Suo J, Chen J, Liu X, Zhao X. (2016). Chronic exposure
mouse models of insulin resistance. PLoS Biology 6(2): e37. to aluminum and risk of Alzheimer’s disease: a meta-analysis. Neurosci Lett
610: 200–206.
Teraoka H. (1981). Distribution of 24 elements in the internal organs of nor-
mal males and the metallic workers in Japan. Arch Environ Health 36: 155– Warady BA, Ford DM, Gaston CE, Sedrnan AB, Huffer WE, Lum GM. (1986).
164. Aluminum intoxication in a child: treatment with intraperitoneal desferri-
oxamine. Pediatrics 78: 651–655.
Thompson KH, Orvig C. (2006). Vanadium in diabetes: 100 years from phase 0
to phase I. J Inorg Biochem 100: 1925–1935. Ward R, Zhang Y, Crichton RR. (2001). Aluminum toxicity and iron homeosta-
sis. J Inorg Biochem 87: 9–14.
Thomson SM, Burnet DC, Bergmann JD, Hixson CJ. (1986). Comparative inha-
lation hazards of aluminum and brass powders using bronchopulmonary Wei H, Wang MD, Meng LX. (2012). Laurel West aluminum industial base resi-
lavage as an indicator of lung damage J Appl Toxicol 6: 197–209. dents trace element content in serum and its associated abnormal glucose
metabolism (J). Xian dai Yu fang Yi xue (Modern Prev Med) 4: 065.
Tomasello MF, Sinopoli A, Pappalardo G. (2015). On the environmental fac-
tors affecting the structural and cytotoxic properties of IAPP peptides. J Di- Wei X, Wei H, Yang D, Li D, Yang X, He M, Lin E, Wu B. (2018). Effect of alumi-
num exposure on glucose metabolism and its mechanism in rats. Biol Trace
abetes Res 2015: 918573.
Elem Res https: //doi.org/10.1007/s12011-018-1318-x First online 28 March
Tomljenovic L, Shaw CA. (2011). Aluminum vaccine adjuvants: are they safe? 2018.
Curr Med Chem 18: 2630–2637.
Wesdock JC, Arnold IMF. (2014). Occupational and environmental health in
Toyokuni S. (2002). Iron and carcinogenesis: From fenton reaction to target the aluminum industry. J Occup Environ Med 56(5 suppl): S5–11.
genes. Redox Rep 4: 189–197.
Westermark P, Andersson A, Westermark GT. (2011). Islet amyloid polypep-
Trieff NM, Romana LA, Esposito A, Oral R, Quiniou F, Iaccarino M, Alcock N, tide, islet amyloid and diabetes mellitus. Physiol Rev 91(3): 795–826.
Ramanujam VMS, Pagano G. (1995). Effluent from bauxite factory induces
Willhite CC, Karyakina NA, Yokel RA, Yenugadhati N, Wisniewski TM, Arnold
developmental and reproductive damage in sea urchins. Arch Environ Con- IMF, Momoli F, Krewski D. (2014). Systematic review of potential health risk
tam Toxicol 28: 173–177. posed by pharmaceutical, occupational and consumer exposures to metal-
Türkez H, Geyikoglu F, Colak S. (2011). The protective effect of boric acid in lic and nanoscale aluminium, aluminum oxides, aluminum hydroxides and
aluminum-induced hepatotoxicity and genotoxicity in rats. Turk J Biol 35: its soluble salts. Crit Rev Toxicol 44(suppl 4): 1–88.
293–301. Wills MR, Savory J. (1983). Aluminum poisoning: Dialysis encephalopathy, os-
Türkez H, Yousef MI, Geyikoglu F. (2010). Propolis prevents aluminium-in- teomalacia and anaemia. Lancet 322: 29–34.
duced genetic and hepatic damages in rat liver. Food Chem Toxicol 48(10): Wills MR, Savory J. (1989). Aluminum and chronic renal failure: Sources, ab-
2741–2746. sorption, transport, and toxicity. Crit Rev Clin Lab Sci 27: 59–107.
Valkonen S Aitio A. (1997). Analysis of aluminum in serum and urine for the Wisniewski HM, Sturman JA, Shek JW. (1980). Aluminum chloride induced
biomonitoring of occupational exposure. Sci Total Environ 199: 103–110. neurofibrillary changes in the developing rabbit: a chronic animal model.
van Landeghem GF, D’Haese PC, Lamberts LV, DebBroe ME. (1994). Quanti- Ann Neurol 8: 479–490.
tative HPLC/ETAAS hybrid method with an on-line metal scavenger for Wisser LA, Heinrich BS, Leach RM. (1990). Effect of aluminum on performance
studying the protein binding and speciation of aluminum and iron. Anal and mineral metabolism in young chicks and laying hens. J Nutr 120(5):
Chem 66: 216–222. 493–498.
van Rensburg SJ, Carstens ME, Potocnik FCV, Aucamp AK, Taljaard JJF, Koch Woodson GC. (1998). An interesting case of osteomalacia due to antacid use
KR. (1992). Membrane fluidity of platelets and erythrocytes in patients associated with stainable bone aluminum in a patient with normal renal
with Alzheimer’s disease and the effect of small amounts of aluminium on function. Bone 22: 695–698.
platelet and erythrocyte membranes. Neurochem Res 17(8): 825–829. Xiea CX, Mattsonbc MP, Lovellcd MA, Yokelae RA. (1996). Intraneuronal alu-
Vandenplas O, Delwiche JP, Vanbilsen ML, Joly J, Roosels D. (1998). Occupa- minum potentiates iron-induced oxidative stress in cultured rat hippocam-
tional asthma caused by aluminum welding. Eur Respir J 11: 1182–1184. pal neurons. Brain Res 743(1–2): 271–277.
Vasanthan S, Joshi P. (2018). Effect of aluminum toxicity and Bacopa monnieri Xu F, Liu Y, Zhao H, Yu K, Song M, Zhu Y, Li Y. (2017). Aluminum chloride
on plasma cortisol level in Wistar albino rats. Natl J Physiol Pharm Pharma- caused liver dysfunction and mitochondrial energy metabolism disorder in
col 8(8): 1088–1091. rat. J Inorg Biochem 174: 55–66.

Copyright © 2019 SETOX & Institute of Experimental Pharmacology and Toxicology, CEM SASc.
70 Aluminium toxicosis
Ikechukwu Onyebuchi Igbokwe, Ephraim Igwenagu, Nanacha Afifi Igbokwe

Xu F, Ren L, Song M, Shao B, Han Y, Cao Z, Li Y. (2018). Fas- and mitochondia- Yousef MI, Salama AF. (2009). Propolis protection from reproductive toxic-
mediated signaling pathway involved in osteoblast apoptosis induced by ity caused by aluminium chloride in male rats. Food Chem Toxicol 47: 1168–
AlCl3. Biol Trace Elem Res 184(1): 173–185. 1175.
Xu ZX, Fox L, Melethil S, Winberg L, Badr M. (1990). Mechanism of aluminum- Yu H, Zhang J, Ji, Q, Wang P, Song M, Cao Z, Zhang X, Li Y. (2019). Melatonin al-
induced inhibition of hepatic glycolysis: inactivation of phosphofructoki- leviates aluminium chloride-induced immunotoxicity by inhibiting oxida-
nase. J Pharmacol Exp Ther 254(1): 301–305. tive stress and apoptosis associated with the activation of Nrf2 signaling
Xu Z-X, Zhang Q, Ma G-L, Chen C-H, He Y-M, Xu L-H, Zhang Y, Zhou G-R, Li pathway. Ecotoxicol Environ Saf 173: 131–141.
Z-H, Yang H-J, Zhou P. (2016). Influence of aluinium and EGCCG on fibrilla- Zatta P, Lain E, Cagnolini C. (2000). Effects of aluminum on activity of Krebs
tion and aggregation of human islet amyloid polypeptide. J Diabetes Res cycle enzymes and glutamate dehydrogenase in rat brain homogenate.
2016, ID 1867059, 14 pages. http: //dx.org/10.1155/2016/1867059. Eur J Biochem 267(10): 3049–3055.
Yahaya T, Obaroh I, Oladele EO. (2017). The roles of environmental pollutants Zatta P, Perazzolo M, Corain B. (1989). Tris acetylacetonate aluminium (III) in-
in the pathogenesis and prevalence of diabetes: a review. J Appl Sci Environ duces osmotic fragility and acanthocyte formation in suspended erythro-
Manag 21: 5–8. cytes. Toxicol Lett 45(1): 15–21.
Yamanaka K, Minato N, Iwai K. (1999). Stabilization of iron regulatory protein Zatta P, Ricchelli F, Drago D, Filipi B, Tognon G. (2005). Aluminum-triggered
2, IRP2, by aluminum. FEBS lett 462: 216–220. structural modification and aggregation of beta-amyloid. Cell Mol Life Sci
Yang AM, ChengN, Pu HQ, Liu SM, Li JS, Bassig BA, Dai M, Li HY, Hu XB, Wei 62: 1725–1733.
X, Zheng TZ, Bai YN. (2015). Metal exposure and risk of diabetes and pre- Zatta P, Zambenedetti P, Milanese M. (1999). Activation of monoamine ox-
diabetes among Chinese occupational workers. Biomed Eviron Sci 28(12): idase type B by aluminum in rat brain homogenate. NeuroReport 10(17):
875–883. 3645–3648.
Yang M, Jiang L, Huang H, Zeng S, Qiu F, Yu , Li X, Wei S. (2014). Dietary ex- Zhang F, Sun X, Yu H, Yang X, Song M, Han Y, Li Y, Zhu Y. (2017). Effects of alu-
posure to aluminium and health risk assessment in residents of Shenzhen, minum trichloride on cartilage stimulatory growth factor in rats. Biometals
China. PLoS One 9(3): e89715 https: //doi.org/10.1371/journalpone.0089715. 30(1): 143–150.
Yang S-J, Huh J-E, Lee JE, Choi SY, Kim TU, Cho S-W. (2003). Inactivation of hu- Zhang L, Lin X, Gu Q, Zhu Y, Zhao H, Li Y, Zhang Z. (2010). Effects of sub-
man glutamate dehydrogenase by aluminum. Cell Mol Life Sci 60(11): 2538– chronic aluminum exposure on serum concentrations of iron and iron-as-
2546. sociated proteins in rats. Biol Trace Elem Res 141(1–3): 246–253.
Yang X, Huo H, Xiu C, Song M, Han Y, Li Y, Zhu Y. (2016). Inhibition of osteo- Zhang Q, Cao Z, Sun X, Zuang C, Huang W, Li Y. (2016). Aluminum trichloride
blast differentiation by aluminum trichloride exposure is associated with induces hypertension and disturbs the function of erythrocyte membrane
inhibition of BMP-2/Smad pathway component expression. Food Chem in male rats. Biol Trace Elem Res 171(1): 116–123.
Toxicol 97: 120–126. Zhang Y, Song W. (2017). Islet amyloid polypeptide: another key molecule in
Yang X, Yu K, Wang H, Zhang H, Bai C, Song M, Han Y, Shao B, Li Y, Li X. (2018). Alzheimer’s pathogenesis? Prog Neurobiol 153: 100–120.
Bone impairment caused AlCl3 is associated with activation of JNK apop- Zhou Y, Harris WR, Yokel RA. (2008). The influence of citrate, maltolate and
totic pathway mediated by oxidative stress. Food Chem Toxicol 116(Part B): fluoride on the gastrointestinal absorption of aluminum at a drinking wa-
307–314. ter-relevant concentration: A 26Al and 14C study. J Inorg Biochem 102: 798–
Yassa HA, George SM, Mohamed HK. (2017). Folic acid improves develop- 808.
mental toxicity induced by aluminum sulphates. Environ Toxicol Pharmacol Zhou Y, Yokel RA. (2005). The chemical species of aluminum influence its
50: 32–36. paracellular flux across and uptake into Caco-2 cells, a model of gastroin-
Yokel RA. (2000). The toxicology of aluminum in the brain: a review. Neuro- testinal absorption. Toxicol Sci 87: 15–26.
toxicology 21: 813–828. Zhu Y, Hu C, Zheng P, Miao L, Yan X, Li H, Wang Z, Gao B, Li Y. (2016a). Ginsen-
Yokel RA. (2012). Aluminum in food- The nature and contributions of food oside Rb1 alleviates aluminum chloride induced rat osteoblasts dysfunc-
additives. Food Additive (El-Samragy Y ed), ISBN: 978-953-51-0067-6, InTech, tion. Toxicology 368–369: 183–188.
http: //www.intechopen.com. Zhu Y, Li Y, Miao L, Wang Y, Liu Y, Yan X. (2014a). Immunotoxicity of alumi-
Yokel RA, Florence RL. (2006). Aluminum bioavailability from the approved num. Chemosphere 104: 1–6.
food additive leavening agent acidic sodium aluminum phosphate, incor- Zhu Y, Xu F, Yan X, Miao L, Li H, Hu C, Wang Z, Lian S, Feng Z, Li Y. (2016b). The
porated into a baked good, is lower than from water. Toxicology 227: 86– suppressive effects of aluminum chloride on osteoblasts function. Environ
93. Toxicol Pharmacol 48: 125–129.
Yokel RA, Hicks CL, Florence RL. (2008). Aluminum bioavailability from ba- Zhu YZ, Sun H, Fu Y, Wang J, Song M, Li M, Li YF, Miao LG. (2014b). Effects of
sic sodium aluminum phosphate, an approved food additive emulsifying sub-chronic aluminium chloride on spermatogenesis and testicular enzy-
agent, incorporated in cheese. Food Chem Toxicol 46: 2261–2266. matic activity in male rats. Life Sci 102(1): 36–40.
Yokel RA, McNamara PJ. (2001). Aluminum toxicokinetics: an updated mini- Zhuang C, Liu D, Yang X, Wang H, Han L, Li Y. (2016). The immunotoxicity of
review. Pharmacol Toxicol 88: 159–167. aluminum trichloride on rat peritoneal macrophages via β2-adrenoceptors/
Yousef MI, Kamel KL, ElGuendi MI, El-Demerdash FM. (2007). An in vitro study cAMP pathway acted by norepinephrine. Chemosphere 149: 34–40.
on reproductive toxicity of aluminium chloride on rabbit sperm: The pro- Zlatkovic J, Tsouchnikas G, Jarmer J, Koessi C, Stiasny K, Heinz FX. (2013). Alu-
tective role of some antioxidants. Toxicology 239: 213–223. minum hydroxide influences not only the extent but also the fine specific-
ity and function activity of antibody responses to tick-borne encephalitis
virus in mice. J Virol 87(22): 12187–12195.

ISSN: 1337-6853 (print version) | 1337-9569 (electronic version)

You might also like